Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Genom ; : 100565, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38781966

ABSTRACT

Spatially resolved transcriptomics (SRT) technologies have revolutionized the study of tissue organization. We introduce a graph convolutional network with an attention and positive emphasis mechanism, termed BINARY, relying exclusively on binarized SRT data to accurately delineate spatial domains. BINARY outperforms existing methods across various SRT data types while using significantly less input information. Our study suggests that precise gene expression quantification may not always be essential, inspiring further exploration of the broader applications of spatially resolved binarized gene expression data.

2.
Nat Methods ; 21(4): 712-722, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38491270

ABSTRACT

Spatial clustering, which shares an analogy with single-cell clustering, has expanded the scope of tissue physiology studies from cell-centroid to structure-centroid with spatially resolved transcriptomics (SRT) data. Computational methods have undergone remarkable development in recent years, but a comprehensive benchmark study is still lacking. Here we present a benchmark study of 13 computational methods on 34 SRT data (7 datasets). The performance was evaluated on the basis of accuracy, spatial continuity, marker genes detection, scalability, and robustness. We found existing methods were complementary in terms of their performance and functionality, and we provide guidance for selecting appropriate methods for given scenarios. On testing additional 22 challenging datasets, we identified challenges in identifying noncontinuous spatial domains and limitations of existing methods, highlighting their inadequacies in handling recent large-scale tasks. Furthermore, with 145 simulated data, we examined the robustness of these methods against four different factors, and assessed the impact of pre- and postprocessing approaches. Our study offers a comprehensive evaluation of existing spatial clustering methods with SRT data, paving the way for future advancements in this rapidly evolving field.


Subject(s)
Benchmarking , Gene Expression Profiling , Cluster Analysis , Spatial Analysis , Transcriptome
3.
Nat Commun ; 15(1): 207, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38182575

ABSTRACT

Tissue structure identification is a crucial task in spatial omics data analysis, for which increasingly complex models, such as Graph Neural Networks and Bayesian networks, are employed. However, whether increased model complexity can effectively lead to improved performance is a notable question in the field. Inspired by the consistent observation of cellular neighborhood structures across various spatial technologies, we propose Multi-range cEll coNtext DEciphereR (MENDER), for tissue structure identification. Applied on datasets of 3 brain regions and a whole-brain atlas, MENDER, with biology-driven design, offers substantial improvements over modern complex models while automatically aligning labels across slices, despite using much less running time than the second-fastest. MENDER's identification power allows the uncovering of previously overlooked spatial domains that exhibit strong associations with brain aging. MENDER's scalability makes it freely appliable on a million-level brain spatial atlas. MENDER's discriminative power enables the differentiation of breast cancer patient subtypes obscured by single-cell analysis.


Subject(s)
Aging , Breast Neoplasms , Humans , Female , Bayes Theorem , Brain , Breast , Breast Neoplasms/genetics
4.
Nat Protoc ; 19(3): 831-895, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38135744

ABSTRACT

Advances in spatial omics technologies have improved the understanding of cellular organization in tissues, leading to the generation of complex and heterogeneous data and prompting the development of specialized tools for managing, loading and visualizing spatial omics data. The Spatial Omics Database (SODB) was established to offer a unified format for data storage and interactive visualization modules. Here we detail the use of Pysodb, a Python-based tool designed to enable the efficient exploration and loading of spatial datasets from SODB within a Python environment. We present seven case studies using Pysodb, detailing the interaction with various computational methods, ensuring reproducibility of experimental data and facilitating the integration of new data and alternative applications in SODB. The approach offers a reference for method developers by outlining label and metadata availability in representative spatial data that can be loaded by Pysodb. The tool is supplemented by a website ( https://protocols-pysodb.readthedocs.io/ ) with detailed information for benchmarking analysis, and allows method developers to focus on computational models by facilitating data processing. This protocol is designed for researchers with limited experience in computational biology. Depending on the dataset complexity, the protocol typically requires ~12 h to complete.


Subject(s)
Computational Biology , Software , Reproducibility of Results , Computational Biology/methods , Databases, Factual , Data Analysis
5.
Genome Biol ; 24(1): 241, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37864231

ABSTRACT

Properly integrating spatially resolved transcriptomics (SRT) generated from different batches into a unified gene-spatial coordinate system could enable the construction of a comprehensive spatial transcriptome atlas. Here, we propose SPIRAL, consisting of two consecutive modules: SPIRAL-integration, with graph domain adaptation-based data integration, and SPIRAL-alignment, with cluster-aware optimal transport-based coordination alignment. We verify SPIRAL with both synthetic and real SRT datasets. By encoding spatial correlations to gene expressions, SPIRAL-integration surpasses state-of-the-art methods in both batch effect removal and joint spatial domain identification. By aligning spots cluster-wise, SPIRAL-alignment achieves more accurate coordinate alignments than existing methods.


Subject(s)
Gene Expression Profiling , Transcriptome
6.
Semin Cancer Biol ; 95: 25-41, 2023 10.
Article in English | MEDLINE | ID: mdl-37400044

ABSTRACT

Spatially resolved transcriptomics (SRT) has unlocked new dimensions in our understanding of intricate tissue architectures. However, this rapidly expanding field produces a wealth of diverse and voluminous data, necessitating the evolution of sophisticated computational strategies to unravel inherent patterns. Two distinct methodologies, gene spatial pattern recognition (GSPR) and tissue spatial pattern recognition (TSPR), have emerged as vital tools in this process. GSPR methodologies are designed to identify and classify genes exhibiting noteworthy spatial patterns, while TSPR strategies aim to understand intercellular interactions and recognize tissue domains with molecular and spatial coherence. In this review, we provide a comprehensive exploration of SRT, highlighting crucial data modalities and resources that are instrumental for the development of methods and biological insights. We address the complexities and challenges posed by the use of heterogeneous data in developing GSPR and TSPR methodologies and propose an optimal workflow for both. We delve into the latest advancements in GSPR and TSPR, examining their interrelationships. Lastly, we peer into the future, envisaging the potential directions and perspectives in this dynamic field.


Subject(s)
Biology , Gene Expression Profiling , Humans , Workflow , Computational Biology , Transcriptome
8.
Nat Methods ; 20(3): 387-399, 2023 03.
Article in English | MEDLINE | ID: mdl-36797409

ABSTRACT

Spatial omics technologies generate wealthy but highly complex datasets. Here we present Spatial Omics DataBase (SODB), a web-based platform providing both rich data resources and a suite of interactive data analytical modules. SODB currently maintains >2,400 experiments from >25 spatial omics technologies, which are freely accessible as a unified data format compatible with various computational packages. SODB also provides multiple interactive data analytical modules, especially a unique module, Spatial Omics View (SOView). We conduct comprehensive statistical analyses and illustrate the utility of both basic and advanced analytical modules using multiple spatial omics datasets. We demonstrate SOView utility with brain spatial transcriptomics data and recover known anatomical structures. We further delineate functional tissue domains with associated marker genes that were obscured when analyzed using previous methods. We finally show how SODB may efficiently facilitate computational method development. The SODB website is https://gene.ai.tencent.com/SpatialOmics/ . The command-line package is available at https://pysodb.readthedocs.io/en/latest/ .


Subject(s)
Gene Expression Profiling , Software , Databases, Factual , Gene Expression Profiling/methods
9.
Nat Commun ; 13(1): 7640, 2022 12 10.
Article in English | MEDLINE | ID: mdl-36496406

ABSTRACT

Spatially resolved transcriptomics provides the opportunity to investigate the gene expression profiles and the spatial context of cells in naive state, but at low transcript detection sensitivity or with limited gene throughput. Comprehensive annotating of cell types in spatially resolved transcriptomics to understand biological processes at the single cell level remains challenging. Here we propose Spatial-ID, a supervision-based cell typing method, that combines the existing knowledge of reference single-cell RNA-seq data and the spatial information of spatially resolved transcriptomics data. We present a series of benchmarking analyses on publicly available spatially resolved transcriptomics datasets, that demonstrate the superiority of Spatial-ID compared with state-of-the-art methods. Besides, we apply Spatial-ID on a self-collected mouse brain hemisphere dataset measured by Stereo-seq, that shows the scalability of Spatial-ID to three-dimensional large field tissues with subcellular spatial resolution.


Subject(s)
Gene Expression Profiling , Single-Cell Analysis , Mice , Animals , Single-Cell Analysis/methods , Gene Expression Profiling/methods , Transcriptome , Intracellular Space , Machine Learning
10.
Nat Commun ; 13(1): 7330, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36443314

ABSTRACT

The rapidly developing spatial omics generated datasets with diverse scales and modalities. However, most existing methods focus on modeling dynamics of single cells while ignore microenvironments (MEs). Here we present SOTIP (Spatial Omics mulTIPle-task analysis), a versatile method incorporating MEs and their interrelationships into a unified graph. Based on this graph, spatial heterogeneity quantification, spatial domain identification, differential microenvironment analysis, and other downstream tasks can be performed. We validate each module's accuracy, robustness, scalability and interpretability on various spatial omics datasets. In two independent mouse cerebral cortex spatial transcriptomics datasets, we reveal a gradient spatial heterogeneity pattern strongly correlated with the cortical depth. In human triple-negative breast cancer spatial proteomics datasets, we identify molecular polarizations and MEs associated with different patient survivals. Overall, by modeling biologically explainable MEs, SOTIP outperforms state-of-art methods and provides some perspectives for spatial omics data exploration and interpretation.


Subject(s)
Cerebral Cortex , Space Flight , Animals , Mice , Humans , Proteomics , Spatial Analysis , Survival
11.
Nucleic Acids Res ; 50(1): 46-56, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34850940

ABSTRACT

Clustering cells and depicting the lineage relationship among cell subpopulations are fundamental tasks in single-cell omics studies. However, existing analytical methods face challenges in stratifying cells, tracking cellular trajectories, and identifying critical points of cell transitions. To overcome these, we proposed a novel Markov hierarchical clustering algorithm (MarkovHC), a topological clustering method that leverages the metastability of exponentially perturbed Markov chains for systematically reconstructing the cellular landscape. Briefly, MarkovHC starts with local connectivity and density derived from the input and outputs a hierarchical structure for the data. We firstly benchmarked MarkovHC on five simulated datasets and ten public single-cell datasets with known labels. Then, we used MarkovHC to investigate the multi-level architectures and transition processes during human embryo preimplantation development and gastric cancer procession. MarkovHC found heterogeneous cell states and sub-cell types in lineage-specific progenitor cells and revealed the most possible transition paths and critical points in the cellular processes. These results demonstrated MarkovHC's effectiveness in facilitating the stratification of cells, identification of cell populations, and characterization of cellular trajectories and critical points.


Subject(s)
Computational Biology/methods , Single-Cell Analysis/methods , Blastocyst/cytology , Blastocyst/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Lineage , Humans , Markov Chains
12.
Nat Methods ; 18(10): 1223-1232, 2021 10.
Article in English | MEDLINE | ID: mdl-34608315

ABSTRACT

Spatial metabolomics can reveal intercellular heterogeneity and tissue organization. Here we report on the spatial single nuclear metabolomics (SEAM) method, a flexible platform combining high-spatial-resolution imaging mass spectrometry and a set of computational algorithms that can display multiscale and multicolor tissue tomography together with identification and clustering of single nuclei by their in situ metabolic fingerprints. We first applied SEAM to a range of wild-type mouse tissues, then delineated a consistent pattern of metabolic zonation in mouse liver. We further studied the spatial metabolic profile in the human fibrotic liver. We discovered subpopulations of hepatocytes with special metabolic features associated with their proximity to the fibrotic niche, and validated this finding by spatial transcriptomics with Geo-seq. These demonstrations highlighted SEAM's ability to explore the spatial metabolic profile and tissue histology at the single-cell level, leading to a deeper understanding of tissue metabolic organization.


Subject(s)
Cellular Microenvironment , Computational Biology/methods , Liver Cirrhosis/metabolism , Liver/cytology , Algorithms , Animals , Hepatocytes/physiology , Humans , Liver/physiology , Metabolomics/methods , Mice , Reproducibility of Results , Single Molecule Imaging , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...