Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Res Sq ; 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38645152

RESUMEN

With the growing number of single-cell analysis tools, benchmarks are increasingly important to guide analysis and method development. However, a lack of standardisation and extensibility in current benchmarks limits their usability, longevity, and relevance to the community. We present Open Problems, a living, extensible, community-guided benchmarking platform including 10 current single-cell tasks that we envision will raise standards for the selection, evaluation, and development of methods in single-cell analysis.

2.
Nat Methods ; 21(4): 673-679, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38438615

RESUMEN

Spatial landmarks are crucial in describing histological features between samples or sites, tracking regions of interest in microscopy, and registering tissue samples within a common coordinate framework. Although other studies have explored unsupervised landmark detection, existing methods are not well-suited for histological image data as they often require a large number of images to converge, are unable to handle nonlinear deformations between tissue sections and are ineffective for z-stack alignment, other modalities beyond image data or multimodal data. We address these challenges by introducing effortless landmark detection, a new unsupervised landmark detection and registration method using neural-network-guided thin-plate splines. Our proposed method is evaluated on a diverse range of datasets including histology and spatially resolved transcriptomics, demonstrating superior performance in both accuracy and stability compared to existing approaches.


Asunto(s)
Aprendizaje Profundo , Procesamiento de Imagen Asistido por Computador/métodos
3.
Science ; 382(6675): eadf8486, 2023 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-38060664

RESUMEN

The spatial distribution of lymphocyte clones within tissues is critical to their development, selection, and expansion. We have developed spatial transcriptomics of variable, diversity, and joining (VDJ) sequences (Spatial VDJ), a method that maps B cell and T cell receptor sequences in human tissue sections. Spatial VDJ captures lymphocyte clones that match canonical B and T cell distributions and amplifies clonal sequences confirmed by orthogonal methods. We found spatial congruency between paired receptor chains, developed a computational framework to predict receptor pairs, and linked the expansion of distinct B cell clones to different tumor-associated gene expression programs. Spatial VDJ delineates B cell clonal diversity and lineage trajectories within their anatomical niche. Thus, Spatial VDJ captures lymphocyte spatial clonal architecture across tissues, providing a platform to harness clonal sequences for therapy.


Asunto(s)
Linfocitos B , Receptores de Células Precursoras de Linfocitos B , Receptores de Antígenos de Linfocitos T , Linfocitos T , Humanos , Linfocitos B/metabolismo , Células Clonales/metabolismo , Perfilación de la Expresión Génica/métodos , Receptores de Células Precursoras de Linfocitos B/genética , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/metabolismo
4.
J Invest Dermatol ; 143(11): 2177-2192.e13, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37142187

RESUMEN

Epidermal homeostasis is governed by a balance between keratinocyte proliferation and differentiation with contributions from cell-cell interactions, but conserved or divergent mechanisms governing this equilibrium across species and how an imbalance contributes to skin disease are largely undefined. To address these questions, human skin single-cell RNA sequencing and spatial transcriptomics data were integrated and compared with mouse skin data. Human skin cell-type annotation was improved using matched spatial transcriptomics data, highlighting the importance of spatial context in cell-type identity, and spatial transcriptomics refined cellular communication inference. In cross-species analyses, we identified a human spinous keratinocyte subpopulation that exhibited proliferative capacity and a heavy metal processing signature, which was absent in mouse and may account for species differences in epidermal thickness. This human subpopulation was expanded in psoriasis and zinc-deficiency dermatitis, attesting to disease relevance and suggesting a paradigm of subpopulation dysfunction as a hallmark of the disease. To assess additional potential subpopulation drivers of skin diseases, we performed cell-of-origin enrichment analysis within genodermatoses, nominating pathogenic cell subpopulations and their communication pathways, which highlighted multiple potential therapeutic targets. This integrated dataset is encompassed in a publicly available web resource to aid mechanistic and translational studies of normal and diseased skin.


Asunto(s)
Enfermedades de la Piel , Transcriptoma , Humanos , Animales , Ratones , Piel , Queratinocitos/metabolismo , Epidermis/patología , Enfermedades de la Piel/patología , Comunicación Celular
5.
Nat Neurosci ; 26(5): 891-901, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37095395

RESUMEN

The spatiotemporal regulation of cell fate specification in the human developing spinal cord remains largely unknown. In this study, by performing integrated analysis of single-cell and spatial multi-omics data, we used 16 prenatal human samples to create a comprehensive developmental cell atlas of the spinal cord during post-conceptional weeks 5-12. This revealed how the cell fate commitment of neural progenitor cells and their spatial positioning are spatiotemporally regulated by specific gene sets. We identified unique events in human spinal cord development relative to rodents, including earlier quiescence of active neural stem cells, differential regulation of cell differentiation and distinct spatiotemporal genetic regulation of cell fate choices. In addition, by integrating our atlas with pediatric ependymomas data, we identified specific molecular signatures and lineage-specific genes of cancer stem cells during progression. Thus, we delineate spatiotemporal genetic regulation of human spinal cord development and leverage these data to gain disease insight.


Asunto(s)
Ependimoma , Células-Madre Neurales , Niño , Femenino , Embarazo , Humanos , Médula Espinal , Ependimoma/genética , Ependimoma/metabolismo , Diferenciación Celular/genética , Células-Madre Neurales/fisiología , Expresión Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética
6.
Nat Commun ; 14(1): 1438, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36922516

RESUMEN

To date, single-cell studies of human white adipose tissue (WAT) have been based on small cohort sizes and no cellular consensus nomenclature exists. Herein, we performed a comprehensive meta-analysis of publicly available and newly generated single-cell, single-nucleus, and spatial transcriptomic results from human subcutaneous, omental, and perivascular WAT. Our high-resolution map is built on data from ten studies and allowed us to robustly identify >60 subpopulations of adipocytes, fibroblast and adipogenic progenitors, vascular, and immune cells. Using these results, we deconvolved spatial and bulk transcriptomic data from nine additional cohorts to provide spatial and clinical dimensions to the map. This identified cell-cell interactions as well as relationships between specific cell subtypes and insulin resistance, dyslipidemia, adipocyte volume, and lipolysis upon long-term weight changes. Altogether, our meta-map provides a rich resource defining the cellular and microarchitectural landscape of human WAT and describes the associations between specific cell types and metabolic states.


Asunto(s)
Tejido Adiposo Blanco , Transcriptoma , Humanos , Transcriptoma/genética , Tejido Adiposo Blanco/metabolismo , Adipocitos/metabolismo , Perfilación de la Expresión Génica , Adipogénesis/genética , Tejido Adiposo
7.
PLoS Comput Biol ; 18(10): e1010583, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36206305

RESUMEN

Calmodulin (CaM) is a calcium sensor which binds and regulates a wide range of target-proteins. This implicitly enables the concentration of calcium to influence many downstream physiological responses, including muscle contraction, learning and depression. The antipsychotic drug trifluoperazine (TFP) is a known CaM inhibitor. By binding to various sites, TFP prevents CaM from associating to target-proteins. However, the molecular and state-dependent mechanisms behind CaM inhibition by drugs such as TFP are largely unknown. Here, we build a Markov state model (MSM) from adaptively sampled molecular dynamics simulations and reveal the structural and dynamical features behind the inhibitory mechanism of TFP-binding to the C-terminal domain of CaM. We specifically identify three major TFP binding-modes from the MSM macrostates, and distinguish their effect on CaM conformation by using a systematic analysis protocol based on biophysical descriptors and tools from machine learning. The results show that depending on the binding orientation, TFP effectively stabilizes features of the calcium-unbound CaM, either affecting the CaM hydrophobic binding pocket, the calcium binding sites or the secondary structure content in the bound domain. The conclusions drawn from this work may in the future serve to formulate a complete model of pharmacological modulation of CaM, which furthers our understanding of how these drugs affect signaling pathways as well as associated diseases.


Asunto(s)
Antipsicóticos , Calmodulina , Calmodulina/metabolismo , Trifluoperazina/farmacología , Trifluoperazina/química , Trifluoperazina/metabolismo , Antipsicóticos/química , Calcio/metabolismo , Unión Proteica , Sitios de Unión
8.
Nature ; 608(7922): 360-367, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35948708

RESUMEN

Defining the transition from benign to malignant tissue is fundamental to improving early diagnosis of cancer1. Here we use a systematic approach to study spatial genome integrity in situ and describe previously unidentified clonal relationships. We used spatially resolved transcriptomics2 to infer spatial copy number variations in >120,000 regions across multiple organs, in benign and malignant tissues. We demonstrate that genome-wide copy number variation reveals distinct clonal patterns within tumours and in nearby benign tissue using an organ-wide approach focused on the prostate. Our results suggest a model for how genomic instability arises in histologically benign tissue that may represent early events in cancer evolution. We highlight the power of capturing the molecular and spatial continuums in a tissue context and challenge the rationale for treatment paradigms, including focal therapy.


Asunto(s)
Células Clonales , Variaciones en el Número de Copia de ADN , Inestabilidad Genómica , Neoplasias , Análisis Espacial , Células Clonales/metabolismo , Células Clonales/patología , Variaciones en el Número de Copia de ADN/genética , Detección Precoz del Cáncer , Genoma Humano , Inestabilidad Genómica/genética , Genómica , Humanos , Masculino , Modelos Biológicos , Neoplasias/genética , Neoplasias/patología , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transcriptoma/genética
9.
Nat Biotechnol ; 40(4): 476-479, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34845373

RESUMEN

Current methods for spatial transcriptomics are limited by low spatial resolution. Here we introduce a method that integrates spatial gene expression data with histological image data from the same tissue section to infer higher-resolution expression maps. Using a deep generative model, our method characterizes the transcriptome of micrometer-scale anatomical features and can predict spatial gene expression from histology images alone.


Asunto(s)
Transcriptoma , Transcriptoma/genética
10.
Nat Commun ; 12(1): 7046, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857782

RESUMEN

Reconstruction of heterogeneity through single cell transcriptional profiling has greatly advanced our understanding of the spatial liver transcriptome in recent years. However, global transcriptional differences across lobular units remain elusive in physical space. Here, we apply Spatial Transcriptomics to perform transcriptomic analysis across sectioned liver tissue. We confirm that the heterogeneity in this complex tissue is predominantly determined by lobular zonation. By introducing novel computational approaches, we enable transcriptional gradient measurements between tissue structures, including several lobules in a variety of orientations. Further, our data suggests the presence of previously transcriptionally uncharacterized structures within liver tissue, contributing to the overall spatial heterogeneity of the organ. This study demonstrates how comprehensive spatial transcriptomic technologies can be used to delineate extensive spatial gene expression patterns in the liver, indicating its future impact for studies of liver function, development and regeneration as well as its potential in pre-clinical and clinical pathology.


Asunto(s)
Heterogeneidad Genética , Hígado/metabolismo , Transcriptoma , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Eritroblastos/citología , Eritroblastos/metabolismo , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Hepatocitos/citología , Hepatocitos/metabolismo , Macrófagos del Hígado/citología , Macrófagos del Hígado/metabolismo , Hígado/citología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Anotación de Secuencia Molecular , Neutrófilos/citología , Neutrófilos/metabolismo
12.
Nat Commun ; 12(1): 6012, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34650042

RESUMEN

In the past decades, transcriptomic studies have revolutionized cancer treatment and diagnosis. However, tumor sequencing strategies typically result in loss of spatial information, critical to understand cell interactions and their functional relevance. To address this, we investigate spatial gene expression in HER2-positive breast tumors using Spatial Transcriptomics technology. We show that expression-based clustering enables data-driven tumor annotation and assessment of intra- and interpatient heterogeneity; from which we discover shared gene signatures for immune and tumor processes. By integration with single cell data, we spatially map tumor-associated cell types to find tertiary lymphoid-like structures, and a type I interferon response overlapping with regions of T-cell and macrophage subset colocalization. We construct a predictive model to infer presence of tertiary lymphoid-like structures, applicable across tissue types and technical platforms. Taken together, we combine different data modalities to define a high resolution map of cellular interactions in tumors and provide tools generalizing across tissues and diseases.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transcriptoma , Neoplasias de la Mama/patología , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Humanos
13.
Nat Genet ; 53(9): 1334-1347, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34493872

RESUMEN

Breast cancers are complex cellular ecosystems where heterotypic interactions play central roles in disease progression and response to therapy. However, our knowledge of their cellular composition and organization is limited. Here we present a single-cell and spatially resolved transcriptomics analysis of human breast cancers. We developed a single-cell method of intrinsic subtype classification (SCSubtype) to reveal recurrent neoplastic cell heterogeneity. Immunophenotyping using cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) provides high-resolution immune profiles, including new PD-L1/PD-L2+ macrophage populations associated with clinical outcome. Mesenchymal cells displayed diverse functions and cell-surface protein expression through differentiation within three major lineages. Stromal-immune niches were spatially organized in tumors, offering insights into antitumor immune regulation. Using single-cell signatures, we deconvoluted large breast cancer cohorts to stratify them into nine clusters, termed 'ecotypes', with unique cellular compositions and clinical outcomes. This study provides a comprehensive transcriptional atlas of the cellular architecture of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Análisis de la Célula Individual , Transcriptoma/genética , Linfocitos B/inmunología , Antígeno B7-H1/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/inmunología , Células Endoteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Macrófagos/citología , Macrófagos/inmunología , Proteínas de la Membrana/genética , Células Mieloides/inmunología , Células Mieloides/metabolismo , Análisis de Secuencia de ARN , Microambiente Tumoral , Proteínas Supresoras de Tumor/genética
14.
Cell Metab ; 33(9): 1869-1882.e6, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34380013

RESUMEN

The contribution of cellular heterogeneity and architecture to white adipose tissue (WAT) function is poorly understood. Herein, we combined spatially resolved transcriptional profiling with single-cell RNA sequencing and image analyses to map human WAT composition and structure. This identified 18 cell classes with unique propensities to form spatially organized homo- and heterotypic clusters. Of these, three constituted mature adipocytes that were similar in size, but distinct in their spatial arrangements and transcriptional profiles. Based on marker genes, we termed these AdipoLEP, AdipoPLIN, and AdipoSAA. We confirmed, in independent datasets, that their respective gene profiles associated differently with both adipocyte and whole-body insulin sensitivity. Corroborating our observations, insulin stimulation in vivo by hyperinsulinemic-euglycemic clamp showed that only AdipoPLIN displayed a transcriptional response to insulin. Altogether, by mining this multimodal resource we identify that human WAT is composed of three classes of mature adipocytes, only one of which is insulin responsive.


Asunto(s)
Resistencia a la Insulina , Insulina , Adipocitos , Tejido Adiposo , Tejido Adiposo Blanco , Humanos , Insulina/farmacología
15.
Bioinformatics ; 37(17): 2644-2650, 2021 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-33704427

RESUMEN

MOTIVATION: Collection of spatial signals in large numbers has become a routine task in multiple omics-fields, but parsing of these rich datasets still pose certain challenges. In whole or near-full transcriptome spatial techniques, spurious expression profiles are intermixed with those exhibiting an organized structure. To distinguish profiles with spatial patterns from the background noise, a metric that enables quantification of spatial structure is desirable. Current methods designed for similar purposes tend to be built around a framework of statistical hypothesis testing, hence we were compelled to explore a fundamentally different strategy. RESULTS: We propose an unexplored approach to analyze spatial transcriptomics data, simulating diffusion of individual transcripts to extract genes with spatial patterns. The method performed as expected when presented with synthetic data. When applied to real data, it identified genes with distinct spatial profiles, involved in key biological processes or characteristic for certain cell types. Compared to existing methods, ours seemed to be less informed by the genes' expression levels and showed better time performance when run with multiple cores. AVAILABILITYAND IMPLEMENTATION: Open-source Python package with a command line interface (CLI), freely available at https://github.com/almaan/sepal under an MIT licence. A mirror of the GitHub repository can be found at Zenodo, doi: 10.5281/zenodo.4573237. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

16.
Cell Genom ; 1(3): 100065, 2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36776149

RESUMEN

Formalin-fixed paraffin embedding (FFPE) is the most widespread long-term tissue preservation approach. Here, we report a procedure to perform genome-wide spatial analysis of mRNA in FFPE-fixed tissue sections, using well-established, commercially available methods for imaging and spatial barcoding using slides spotted with barcoded oligo(dT) probes to capture the 3' end of mRNA molecules in tissue sections. We applied this method for expression profiling and cell type mapping in coronal sections from the mouse brain to demonstrate the method's capability to delineate anatomical regions from a molecular perspective. We also profiled the spatial composition of transcriptomic signatures in two ovarian carcinosarcoma samples, exemplifying the method's potential to elucidate molecular mechanisms in heterogeneous clinical samples. Finally, we demonstrate the applicability of the assay to characterize human lung and kidney organoids and a human lung biopsy specimen infected with SARS-CoV-2. We anticipate that genome-wide spatial gene expression profiling in FFPE biospecimens will be used for retrospective analysis of biobank samples, which will facilitate longitudinal studies of biological processes and biomarker discovery.

17.
Commun Biol ; 3(1): 565, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33037292

RESUMEN

The field of spatial transcriptomics is rapidly expanding, and with it the repertoire of available technologies. However, several of the transcriptome-wide spatial assays do not operate on a single cell level, but rather produce data comprised of contributions from a - potentially heterogeneous - mixture of cells. Still, these techniques are attractive to use when examining complex tissue specimens with diverse cell populations, where complete expression profiles are required to properly capture their richness. Motivated by an interest to put gene expression into context and delineate the spatial arrangement of cell types within a tissue, we here present a model-based probabilistic method that uses single cell data to deconvolve the cell mixtures in spatial data. To illustrate the capacity of our method, we use data from different experimental platforms and spatially map cell types from the mouse brain and developmental heart, which arrange as expected.


Asunto(s)
Biología Computacional , Perfilación de la Expresión Génica , Análisis de la Célula Individual , Transcriptoma , Animales , Biología Computacional/métodos , Biología Computacional/normas , Perfilación de la Expresión Génica/métodos , Humanos , Ratones , Especificidad de Órganos , Organogénesis/genética , Análisis de la Célula Individual/métodos , Análisis de la Célula Individual/normas
18.
Nat Biomed Eng ; 4(8): 827-834, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32572199

RESUMEN

Spatial transcriptomics allows for the measurement of RNA abundance at a high spatial resolution, making it possible to systematically link the morphology of cellular neighbourhoods and spatially localized gene expression. Here, we report the development of a deep learning algorithm for the prediction of local gene expression from haematoxylin-and-eosin-stained histopathology images using a new dataset of 30,612 spatially resolved gene expression data matched to histopathology images from 23 patients with breast cancer. We identified over 100 genes, including known breast cancer biomarkers of intratumoral heterogeneity and the co-localization of tumour growth and immune activation, the expression of which can be predicted from the histopathology images at a resolution of 100 µm. We also show that the algorithm generalizes well to The Cancer Genome Atlas and to other breast cancer gene expression datasets without the need for re-training. Predicting the spatially resolved transcriptome of a tissue directly from tissue images may enable image-based screening for molecular biomarkers with spatial variation.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Aprendizaje Profundo , Algoritmos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Procesamiento de Imagen Asistido por Computador , Reproducibilidad de los Resultados , Transcriptoma
19.
J Membr Biol ; 251(3): 419-430, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29476260

RESUMEN

Viral potassium channels (Kcv) are homologous to the pore module of complex [Formula: see text]-selective ion channels of cellular organisms. Due to their relative simplicity, they have attracted interest towards understanding the principles of [Formula: see text] conduction and channel gating. In this work, we construct a homology model of the [Formula: see text] open state, which we validate by studying the binding of known blockers and by monitoring ion conduction through the channel. Molecular dynamics simulations of this model reveal that the re-orientation of selectivity filter carbonyl groups coincides with the transport of potassium ions, suggesting a possible mechanism for fast gating. In addition, we show that the voltage sensitivity of this mechanism can originate from the relocation of potassium ions inside the selectivity filter. We also explore the interaction of [Formula: see text] with the surrounding bilayer and observe the binding of lipids in the area between two adjacent subunits. The model is available to the scientific community to further explore the structure/function relationship of Kcv channels.


Asunto(s)
Membrana Dobles de Lípidos/química , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Canales de Potasio/química , Canales de Potasio/metabolismo , Potasio/metabolismo , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Simulación de Dinámica Molecular , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...