Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nature ; 616(7955): 113-122, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36922587

RESUMO

Emerging spatial technologies, including spatial transcriptomics and spatial epigenomics, are becoming powerful tools for profiling of cellular states in the tissue context1-5. However, current methods capture only one layer of omics information at a time, precluding the possibility of examining the mechanistic relationship across the central dogma of molecular biology. Here, we present two technologies for spatially resolved, genome-wide, joint profiling of the epigenome and transcriptome by cosequencing chromatin accessibility and gene expression, or histone modifications (H3K27me3, H3K27ac or H3K4me3) and gene expression on the same tissue section at near-single-cell resolution. These were applied to embryonic and juvenile mouse brain, as well as adult human brain, to map how epigenetic mechanisms control transcriptional phenotype and cell dynamics in tissue. Although highly concordant tissue features were identified by either spatial epigenome or spatial transcriptome we also observed distinct patterns, suggesting their differential roles in defining cell states. Linking epigenome to transcriptome pixel by pixel allows the uncovering of new insights in spatial epigenetic priming, differentiation and gene regulation within the tissue architecture. These technologies are of great interest in life science and biomedical research.


Assuntos
Cromatina , Epigenoma , Mamíferos , Transcriptoma , Animais , Humanos , Camundongos , Cromatina/genética , Cromatina/metabolismo , Epigênese Genética , Epigenômica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Mamíferos/genética , Histonas/química , Histonas/metabolismo , Análise de Célula Única , Especificidade de Órgãos , Encéfalo/embriologia , Encéfalo/metabolismo , Envelhecimento/genética
2.
Nat Methods ; 20(11): 1769-1779, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37919419

RESUMO

Recent advancements in single-cell technologies allow characterization of experimental perturbations at single-cell resolution. While methods have been developed to analyze such experiments, the application of a strict causal framework has not yet been explored for the inference of treatment effects at the single-cell level. Here we present a causal-inference-based approach to single-cell perturbation analysis, termed CINEMA-OT (causal independent effect module attribution + optimal transport). CINEMA-OT separates confounding sources of variation from perturbation effects to obtain an optimal transport matching that reflects counterfactual cell pairs. These cell pairs represent causal perturbation responses permitting a number of novel analyses, such as individual treatment-effect analysis, response clustering, attribution analysis, and synergy analysis. We benchmark CINEMA-OT on an array of treatment-effect estimation tasks for several simulated and real datasets and show that it outperforms other single-cell perturbation analysis methods. Finally, we perform CINEMA-OT analysis of two newly generated datasets: (1) rhinovirus and cigarette-smoke-exposed airway organoids, and (2) combinatorial cytokine stimulation of immune cells. In these experiments, CINEMA-OT reveals potential mechanisms by which cigarette-smoke exposure dulls the airway antiviral response, as well as the logic that governs chemokine secretion and peripheral immune cell recruitment.


Assuntos
Citocinas , Filmes Cinematográficos
3.
bioRxiv ; 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38014345

RESUMO

With the emerging single-cell RNA-seq datasets at atlas levels, the potential of a universal model built on existing atlas that can extrapolate to new data remains unclear.A fundamental yet challenging problem for such a model is to identify the underlying biological and batch variations in a zero-shot manner, which is crucial for characterizing scRNA-seq datasets with new biological states. In this work, we present scShift, a mechanistic model that learns batch and biological patterns from atlas-level scRNA-seq data as well as perturbation scRNA-seq data. scShift models genes as functions of latent biological processes, with sparse shifts induced by batch effects and biological perturbations, leveraging recent advances of causal representation learning. Through benchmarking in holdout real datasets, we show scShift reveals unified cell type representations as well as underlying biological variations for query data in zero-shot manners, outperforming widely-used atlas integration, batch correction, and perturbation modeling approaches. scShift enables mapping of gene expression profiles to perturbation labels, and predicts meaningful targets for exhausted T cells as well as a list of diseases in the CellxGene blood atlas.

4.
bioRxiv ; 2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37693629

RESUMO

Spatial omics analyze gene expression and interaction dynamics in relation to tissue structure and function. However, existing methods cannot model the intrinsic and spatial-induced variation in spatial omics data, thus failing to identify true spatial interaction effects. Here, we present Spatial Interaction Modeling using Variational Inference (SIMVI), an annotation-free framework that disentangles cell intrinsic and spatial-induced latent variables for modeling gene expression in spatial omics data. SIMVI enables novel downstream analyses, such as clustering and differential expression analysis based on disentangled representations, spatial effect (SE) identification, SE interpretation, and transfer learning on new measurements / modalities. We benchmarked SIMVI on both simulated and real datasets and show that SIMVI uniquely generates highly accurate SE inferences in synthetic datasets and unveils intrinsic variation in complex real datasets. We applied SIMVI to spatial omics data from diverse platforms and tissues (MERFISH human cortex / mouse liver, Slide-seqv2 mouse hippocampus, Spatial-ATAC-RNA-seq) and revealed various region-specific and cell-type-specific spatial interactions. In addition, our experiments on MERFISH human cortex and spatial-ATAC-RNA-seq showcased SIMVI's power in identifying SEs for new samples / modalities. Finally, we applied SIMVI on a newly collected CosMx melanoma dataset. Using SIMVI, we identified immune cells associated with spatial-dependent interactions and revealed the underlying spatial variations associated with patient outcomes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA