Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38917789

RESUMEN

Spatial transcriptomics (ST) methods unlock molecular mechanisms underlying tissue development, homeostasis, or disease. However, there is a need for easy-to-use, high-resolution, cost-efficient, and 3D-scalable methods. Here, we report Open-ST, a sequencing-based, open-source experimental and computational resource to address these challenges and to study the molecular organization of tissues in 2D and 3D. In mouse brain, Open-ST captured transcripts at subcellular resolution and reconstructed cell types. In primary head-and-neck tumors and patient-matched healthy/metastatic lymph nodes, Open-ST captured the diversity of immune, stromal, and tumor populations in space, validated by imaging-based ST. Distinct cell states were organized around cell-cell communication hotspots in the tumor but not the metastasis. Strikingly, the 3D reconstruction and multimodal analysis of the metastatic lymph node revealed spatially contiguous structures not visible in 2D and potential biomarkers precisely at the 3D tumor/lymph node boundary. All protocols and software are available at https://rajewsky-lab.github.io/openst.

2.
Genome Med ; 14(1): 103, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36085050

RESUMEN

BACKGROUND: Acute kidney injury (AKI) occurs frequently in critically ill patients and is associated with adverse outcomes. Cellular mechanisms underlying AKI and kidney cell responses to injury remain incompletely understood. METHODS: We performed single-nuclei transcriptomics, bulk transcriptomics, molecular imaging studies, and conventional histology on kidney tissues from 8 individuals with severe AKI (stage 2 or 3 according to Kidney Disease: Improving Global Outcomes (KDIGO) criteria). Specimens were obtained within 1-2 h after individuals had succumbed to critical illness associated with respiratory infections, with 4 of 8 individuals diagnosed with COVID-19. Control kidney tissues were obtained post-mortem or after nephrectomy from individuals without AKI. RESULTS: High-depth single cell-resolved gene expression data of human kidneys affected by AKI revealed enrichment of novel injury-associated cell states within the major cell types of the tubular epithelium, in particular in proximal tubules, thick ascending limbs, and distal convoluted tubules. Four distinct, hierarchically interconnected injured cell states were distinguishable and characterized by transcriptome patterns associated with oxidative stress, hypoxia, interferon response, and epithelial-to-mesenchymal transition, respectively. Transcriptome differences between individuals with AKI were driven primarily by the cell type-specific abundance of these four injury subtypes rather than by private molecular responses. AKI-associated changes in gene expression between individuals with and without COVID-19 were similar. CONCLUSIONS: The study provides an extensive resource of the cell type-specific transcriptomic responses associated with critical illness-associated AKI in humans, highlighting recurrent disease-associated signatures and inter-individual heterogeneity. Personalized molecular disease assessment in human AKI may foster the development of tailored therapies.


Asunto(s)
Lesión Renal Aguda , COVID-19 , Lesión Renal Aguda/genética , COVID-19/genética , Enfermedad Crítica , Humanos , Riñón , Transcriptoma
3.
Gigascience ; 112022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35852420

RESUMEN

BACKGROUND: Spatial sequencing methods increasingly gain popularity within RNA biology studies. State-of-the-art techniques quantify messenger RNA expression levels from tissue sections and at the same time register information about the original locations of the molecules in the tissue. The resulting data sets are processed and analyzed by accompanying software that, however, is incompatible across inputs from different technologies. FINDINGS: Here, we present spacemake, a modular, robust, and scalable spatial transcriptomics pipeline built in Snakemake and Python. Spacemake is designed to handle all major spatial transcriptomics data sets and can be readily configured for other technologies. It can process and analyze several samples in parallel, even if they stem from different experimental methods. Spacemake's unified framework enables reproducible data processing from raw sequencing data to automatically generated downstream analysis reports. Spacemake is built with a modular design and offers additional functionality such as sample merging, saturation analysis, and analysis of long reads as separate modules. Moreover, spacemake employs novoSpaRc to integrate spatial and single-cell transcriptomics data, resulting in increased gene counts for the spatial data set. Spacemake is open source and extendable, and it can be seamlessly integrated with existing computational workflows.


Asunto(s)
Programas Informáticos , Transcriptoma , Biología Computacional/métodos , ARN Mensajero , Flujo de Trabajo
4.
NAR Genom Bioinform ; 4(2): lqac042, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35685220

RESUMEN

Advancing technologies that quantify gene expression in space are transforming contemporary biology research. A class of spatial transcriptomics methods uses barcoded bead arrays that are optically decoded via microscopy and are later matched to sequenced data from the respective libraries. To obtain a detailed representation of the tissue in space, robust and efficient computational pipelines are required to process microscopy images and accurately basecall the bead barcodes. Optocoder is a computational framework that processes microscopy images to decode bead barcodes in space. It efficiently aligns images, detects beads, and corrects for confounding factors of the fluorescence signal, such as crosstalk and phasing. Furthermore, Optocoder employs supervised machine learning to strongly increase the number of matches between optically decoded and sequenced barcodes. We benchmark Optocoder using data from an in-house spatial transcriptomics platform, as well as from Slide-Seq(V2), and we show that it efficiently processes all datasets without modification. Optocoder is publicly available, open-source and provided as a stand-alone Python package on GitHub: https://github.com/rajewsky-lab/optocoder.

5.
Nat Protoc ; 16(9): 4177-4200, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34349282

RESUMEN

Single-cell RNA-sequencing (scRNA-seq) technologies have revolutionized modern biomedical sciences. A fundamental challenge is to incorporate spatial information to study tissue organization and spatial gene expression patterns. Here, we describe a detailed protocol for using novoSpaRc, a computational framework that probabilistically assigns cells to tissue locations. At the core of this framework lies a structural correspondence hypothesis, that cells in physical proximity share similar gene expression profiles. Given scRNA-seq data, novoSpaRc spatially reconstructs tissues based on this hypothesis, and optionally, by including a reference atlas of marker genes to improve reconstruction. We describe the novoSpaRc algorithm, and its implementation in an open-source Python package ( https://pypi.org/project/novosparc ). NovoSpaRc maps a scRNA-seq dataset of 10,000 cells onto 1,000 locations in <5 min. We describe results obtained using novoSpaRc to reconstruct the mouse organ of Corti de novo based on the structural correspondence assumption and human osteosarcoma cultured cells based on marker gene information, and provide a step-by-step guide to Drosophila embryo reconstruction in the Procedure to demonstrate how these two strategies can be combined.


Asunto(s)
Expresión Génica , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Programas Informáticos , Análisis Espacial , Algoritmos , Animales , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Humanos , Órgano Espiral/citología , Órgano Espiral/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/patología
6.
Nat Commun ; 12(1): 1929, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33771987

RESUMEN

Leigh syndrome (LS) is a severe manifestation of mitochondrial disease in children and is currently incurable. The lack of effective models hampers our understanding of the mechanisms underlying the neuronal pathology of LS. Using patient-derived induced pluripotent stem cells and CRISPR/Cas9 engineering, we developed a human model of LS caused by mutations in the complex IV assembly gene SURF1. Single-cell RNA-sequencing and multi-omics analysis revealed compromised neuronal morphogenesis in mutant neural cultures and brain organoids. The defects emerged at the level of neural progenitor cells (NPCs), which retained a glycolytic proliferative state that failed to instruct neuronal morphogenesis. LS NPCs carrying mutations in the complex I gene NDUFS4 recapitulated morphogenesis defects. SURF1 gene augmentation and PGC1A induction via bezafibrate treatment supported the metabolic programming of LS NPCs, leading to restored neuronal morphogenesis. Our findings provide mechanistic insights and suggest potential interventional strategies for a rare mitochondrial disease.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Leigh/genética , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Mutación , Neuronas/metabolismo , Organoides/metabolismo , Células Cultivadas , Preescolar , Humanos , Células Madre Pluripotentes Inducidas/citología , Enfermedad de Leigh/metabolismo , Masculino , Metabolómica/métodos , Mitocondrias/genética , Mitocondrias/metabolismo , Morfogénesis/genética , Neuronas/citología , Proteómica/métodos , Análisis de la Célula Individual/métodos , Secuenciación del Exoma
7.
J Am Soc Nephrol ; 32(2): 291-306, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33239393

RESUMEN

BACKGROUND: Single-cell transcriptomes from dissociated tissues provide insights into cell types and their gene expression and may harbor additional information on spatial position and the local microenvironment. The kidney's cells are embedded into a gradient of increasing tissue osmolality from the cortex to the medulla, which may alter their transcriptomes and provide cues for spatial reconstruction. METHODS: Single-cell or single-nuclei mRNA sequencing of dissociated mouse kidneys and of dissected cortex, outer, and inner medulla, to represent the corticomedullary axis, was performed. Computational approaches predicted the spatial ordering of cells along the corticomedullary axis and quantitated expression levels of osmo-responsive genes. In situ hybridization validated computational predictions of spatial gene-expression patterns. The strategy was used to compare single-cell transcriptomes from wild-type mice to those of mice with a collecting duct-specific knockout of the transcription factor grainyhead-like 2 (Grhl2CD-/-), which display reduced renal medullary osmolality. RESULTS: Single-cell transcriptomics from dissociated kidneys provided sufficient information to approximately reconstruct the spatial position of kidney tubule cells and to predict corticomedullary gene expression. Spatial gene expression in the kidney changes gradually and osmo-responsive genes follow the physiologic corticomedullary gradient of tissue osmolality. Single-nuclei transcriptomes from Grhl2CD-/- mice indicated a flattened expression gradient of osmo-responsive genes compared with control mice, consistent with their physiologic phenotype. CONCLUSIONS: Single-cell transcriptomics from dissociated kidneys facilitated the prediction of spatial gene expression along the corticomedullary axis and quantitation of osmotically regulated genes, allowing the prediction of a physiologic phenotype.


Asunto(s)
Corteza Renal/metabolismo , Corteza Renal/patología , Médula Renal/metabolismo , Médula Renal/patología , Transcriptoma , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hibridación in Situ , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Concentración Osmolar
8.
Life Sci Alliance ; 3(11)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32972997

RESUMEN

Single-cell RNA-sequencing (scRNAseq) technologies are rapidly evolving. Although very informative, in standard scRNAseq experiments, the spatial organization of the cells in the tissue of origin is lost. Conversely, spatial RNA-seq technologies designed to maintain cell localization have limited throughput and gene coverage. Mapping scRNAseq to genes with spatial information increases coverage while providing spatial location. However, methods to perform such mapping have not yet been benchmarked. To fill this gap, we organized the DREAM Single-Cell Transcriptomics challenge focused on the spatial reconstruction of cells from the Drosophila embryo from scRNAseq data, leveraging as silver standard, genes with in situ hybridization data from the Berkeley Drosophila Transcription Network Project reference atlas. The 34 participating teams used diverse algorithms for gene selection and location prediction, while being able to correctly localize clusters of cells. Selection of predictor genes was essential for this task. Predictor genes showed a relatively high expression entropy, high spatial clustering and included prominent developmental genes such as gap and pair-rule genes and tissue markers. Application of the top 10 methods to a zebra fish embryo dataset yielded similar performance and statistical properties of the selected genes than in the Drosophila data. This suggests that methods developed in this challenge are able to extract generalizable properties of genes that are useful to accurately reconstruct the spatial arrangement of cells in tissues.


Asunto(s)
Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Análisis de la Célula Individual/métodos , Análisis Espacial , Algoritmos , Animales , Bases de Datos Genéticas , Drosophila/genética , Predicción/métodos , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Análisis de Secuencia de ARN/métodos , Transcriptoma/genética , Pez Cebra/genética
9.
Nat Commun ; 10(1): 5776, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852888

RESUMEN

Skeletal muscle stem cells, called satellite cells and defined by the transcription factor PAX7, are responsible for postnatal muscle growth, homeostasis and regeneration. Attempts to utilize the regenerative potential of muscle stem cells for therapeutic purposes so far failed. We previously established the existence of human PAX7-positive cell colonies with high regenerative potential. We now identified PAX7-negative human muscle-derived cell colonies also positive for the myogenic markers desmin and MYF5. These include cells from a patient with a homozygous PAX7 c.86-1G > A mutation (PAX7null). Single cell and bulk transcriptome analysis show high intra- and inter-donor heterogeneity and reveal the endothelial cell marker CLEC14A to be highly expressed in PAX7null cells. All PAX7-negative cell populations, including PAX7null, form myofibers after transplantation into mice, and regenerate muscle after reinjury. Transplanted PAX7neg cells repopulate the satellite cell niche where they re-express PAX7, or, strikingly, CLEC14A. In conclusion, transplanted human cells do not depend on PAX7 for muscle regeneration.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Lectinas Tipo C/fisiología , Músculo Esquelético/fisiología , Factor de Transcripción PAX7/genética , Regeneración , Células Satélite del Músculo Esquelético/fisiología , Síndrome Debilitante/genética , Animales , Biopsia , Preescolar , Consanguinidad , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Mutación , Factor de Transcripción PAX7/metabolismo , Cultivo Primario de Células , Células Satélite del Músculo Esquelético/trasplante , Análisis de la Célula Individual , Trasplante Heterólogo/métodos , Síndrome Debilitante/terapia , Secuenciación del Exoma
10.
Nature ; 576(7785): 132-137, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31748748

RESUMEN

Multiplexed RNA sequencing in individual cells is transforming basic and clinical life sciences1-4. Often, however, tissues must first be dissociated, and crucial information about spatial relationships and communication between cells is thus lost. Existing approaches to reconstruct tissues assign spatial positions to each cell, independently of other cells, by using spatial patterns of expression of marker genes5,6-which often do not exist. Here we reconstruct spatial positions with little or no prior knowledge, by searching for spatial arrangements of sequenced cells in which nearby cells have transcriptional profiles that are often (but not always) more similar than cells that are farther apart. We formulate this task as a generalized optimal-transport problem for probabilistic embedding and derive an efficient iterative algorithm to solve it. We reconstruct the spatial expression of genes in mammalian liver and intestinal epithelium, fly and zebrafish embryos, sections from the mammalian cerebellum and whole kidney, and use the reconstructed tissues to identify genes that are spatially informative. Thus, we identify an organization principle for the spatial expression of genes in animal tissues, which can be exploited to infer meaningful probabilities of spatial position for individual cells. Our framework ('novoSpaRc') can incorporate prior spatial information and is compatible with any single-cell technology. Additional principles that underlie the cartography of gene expression can be tested using our approach.


Asunto(s)
Expresión Génica , Animales , Drosophila melanogaster , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Programas Informáticos
11.
Nat Methods ; 16(9): 879-886, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31384046

RESUMEN

Although messenger RNAs are key molecules for understanding life, until now, no method has existed to determine the full-length sequence of endogenous mRNAs including their poly(A) tails. Moreover, although non-A nucleotides can be incorporated in poly(A) tails, there also exists no method to accurately sequence them. Here, we present full-length poly(A) and mRNA sequencing (FLAM-seq), a rapid and simple method for high-quality sequencing of entire mRNAs. We report a complementary DNA library preparation method coupled to single-molecule sequencing to perform FLAM-seq. Using human cell lines, brain organoids and Caenorhabditis elegans we show that FLAM-seq delivers high-quality full-length mRNA sequences for thousands of different genes per sample. We find that 3' untranslated region length is correlated with poly(A) tail length, that alternative polyadenylation sites and alternative promoters for the same gene are linked to different tail lengths, and that tails contain a substantial number of cytosines.


Asunto(s)
Encéfalo/metabolismo , Organoides/metabolismo , Poli A/química , Poli A/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN/métodos , Animales , Caenorhabditis elegans , Regulación de la Expresión Génica , Células HeLa , Humanos , Poli A/genética , Poliadenilación , Regiones Promotoras Genéticas , ARN Mensajero/genética
12.
J Am Soc Nephrol ; 29(8): 2060-2068, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29794128

RESUMEN

Background Three different cell types constitute the glomerular filter: mesangial cells, endothelial cells, and podocytes. However, to what extent cellular heterogeneity exists within healthy glomerular cell populations remains unknown.Methods We used nanodroplet-based highly parallel transcriptional profiling to characterize the cellular content of purified wild-type mouse glomeruli.Results Unsupervised clustering of nearly 13,000 single-cell transcriptomes identified the three known glomerular cell types. We provide a comprehensive online atlas of gene expression in glomerular cells that can be queried and visualized using an interactive and freely available database. Novel marker genes for all glomerular cell types were identified and supported by immunohistochemistry images obtained from the Human Protein Atlas. Subclustering of endothelial cells revealed a subset of endothelium that expressed marker genes related to endothelial proliferation. By comparison, the podocyte population appeared more homogeneous but contained three smaller, previously unknown subpopulations.Conclusions Our study comprehensively characterized gene expression in individual glomerular cells and sets the stage for the dissection of glomerular function at the single-cell level in health and disease.


Asunto(s)
Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Glomérulos Renales/fisiología , Células Mesangiales/metabolismo , Podocitos/metabolismo , Análisis de Secuencia de ARN , Animales , Células Cultivadas , Regulación de la Expresión Génica , Glomérulos Renales/citología , Masculino , Ratones , Ratones Endogámicos , Valores de Referencia
13.
Science ; 358(6360): 194-199, 2017 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-28860209

RESUMEN

By the onset of morphogenesis, Drosophila embryos consist of about 6000 cells that express distinct gene combinations. Here, we used single-cell sequencing of precisely staged embryos and devised DistMap, a computational mapping strategy to reconstruct the embryo and to predict spatial gene expression approaching single-cell resolution. We produced a virtual embryo with about 8000 expressed genes per cell. Our interactive Drosophila Virtual Expression eXplorer (DVEX) database generates three-dimensional virtual in situ hybridizations and computes gene expression gradients. We used DVEX to uncover patterned expression of transcription factors and long noncoding RNAs, as well as signaling pathway components. Spatial regulation of Hippo signaling during early embryogenesis suggests a mechanism for establishing asynchronous cell proliferation. Our approach is suitable to generate transcriptomic blueprints for other complex tissues.


Asunto(s)
Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Embrión no Mamífero/citología , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Comunicación Celular , Proteínas de Drosophila/genética , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/genética
14.
BMC Biol ; 15(1): 44, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28526029

RESUMEN

BACKGROUND: Recent developments in droplet-based microfluidics allow the transcriptional profiling of thousands of individual cells in a quantitative, highly parallel and cost-effective way. A critical, often limiting step is the preparation of cells in an unperturbed state, not altered by stress or ageing. Other challenges are rare cells that need to be collected over several days or samples prepared at different times or locations. METHODS: Here, we used chemical fixation to address these problems. Methanol fixation allowed us to stabilise and preserve dissociated cells for weeks without compromising single-cell RNA sequencing data. RESULTS: By using mixtures of fixed, cultured human and mouse cells, we first showed that individual transcriptomes could be confidently assigned to one of the two species. Single-cell gene expression from live and fixed samples correlated well with bulk mRNA-seq data. We then applied methanol fixation to transcriptionally profile primary cells from dissociated, complex tissues. Low RNA content cells from Drosophila embryos, as well as mouse hindbrain and cerebellum cells prepared by fluorescence-activated cell sorting, were successfully analysed after fixation, storage and single-cell droplet RNA-seq. We were able to identify diverse cell populations, including neuronal subtypes. As an additional resource, we provide 'dropbead', an R package for exploratory data analysis, visualization and filtering of Drop-seq data. CONCLUSIONS: We expect that the availability of a simple cell fixation method will open up many new opportunities in diverse biological contexts to analyse transcriptional dynamics at single-cell resolution.


Asunto(s)
Células Cultivadas/citología , Citometría de Flujo/métodos , Perfilación de la Expresión Génica/métodos , Análisis de la Célula Individual/métodos , Animales , Cerebelo/citología , Drosophila/citología , Embrión no Mamífero/citología , Citometría de Flujo/instrumentación , Perfilación de la Expresión Génica/instrumentación , Humanos , Metanol/química , Ratones , ARN Mensajero/análisis , Rombencéfalo/citología , Análisis de Secuencia de ARN , Análisis de la Célula Individual/instrumentación , Programas Informáticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...