Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(2): 375-389.e18, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38242085

RESUMEN

Immune checkpoint inhibition treatment using aPD-1 monoclonal antibodies is a promising cancer immunotherapy approach. However, its effect on tumor immunity is narrow, as most patients do not respond to the treatment or suffer from recurrence. We show that the crosstalk between conventional type I dendritic cells (cDC1) and T cells is essential for an effective aPD-1-mediated anti-tumor response. Accordingly, we developed a bispecific DC-T cell engager (BiCE), a reagent that facilitates physical interactions between PD-1+ T cells and cDC1. BiCE treatment promotes the formation of active dendritic/T cell crosstalk in the tumor and tumor-draining lymph nodes. In vivo, single-cell and physical interacting cell analysis demonstrates the distinct and superior immune reprogramming of the tumors and tumor-draining lymph nodes treated with BiCE as compared to conventional aPD-1 treatment. By bridging immune cells, BiCE potentiates cell circuits and communication pathways needed for effective anti-tumor immunity.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Humanos , Anticuerpos Biespecíficos/uso terapéutico , Células Dendríticas/inmunología , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología
2.
Cell ; 186(12): 2610-2627.e18, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37209682

RESUMEN

The hourglass model describes the convergence of species within the same phylum to a similar body plan during development; however, the molecular mechanisms underlying this phenomenon in mammals remain poorly described. Here, we compare rabbit and mouse time-resolved differentiation trajectories to revisit this model at single-cell resolution. We modeled gastrulation dynamics using hundreds of embryos sampled between gestation days 6.0 and 8.5 and compared the species using a framework for time-resolved single-cell differentiation-flows analysis. We find convergence toward similar cell-state compositions at E7.5, supported by the quantitatively conserved expression of 76 transcription factors, despite divergence in surrounding trophoblast and hypoblast signaling. However, we observed noticeable changes in specification timing of some lineages and divergence of primordial germ cell programs, which in the rabbit do not activate mesoderm genes. Comparative analysis of temporal differentiation models provides a basis for studying the evolution of gastrulation dynamics across mammals.


Asunto(s)
Gastrulación , Mesodermo , Animales , Conejos , Ratones , Gastrulación/genética , Mesodermo/fisiología , Diferenciación Celular/fisiología , Mamíferos/genética , Trofoblastos , Regulación del Desarrollo de la Expresión Génica
3.
Cell ; 185(17): 3169-3185.e20, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35908548

RESUMEN

Mice deficient for all ten-eleven translocation (TET) genes exhibit early gastrulation lethality. However, separating cause and effect in such embryonic failure is challenging. To isolate cell-autonomous effects of TET loss, we used temporal single-cell atlases from embryos with partial or complete mutant contributions. Strikingly, when developing within a wild-type embryo, Tet-mutant cells retain near-complete differentiation potential, whereas embryos solely comprising mutant cells are defective in epiblast to ectoderm transition with degenerated mesoderm potential. We map de-repressions of early epiblast factors (e.g., Dppa4 and Gdf3) and failure to activate multiple signaling from nascent mesoderm (Lefty, FGF, and Notch) as likely cell-intrinsic drivers of TET loss phenotypes. We further suggest loss of enhancer demethylation as the underlying mechanism. Collectively, our work demonstrates an unbiased approach for defining intrinsic and extrinsic embryonic gene function based on temporal differentiation atlases and disentangles the intracellular effects of the demethylation machinery from its broader tissue-level ramifications.


Asunto(s)
Gastrulación , Mesodermo , Animales , Diferenciación Celular/genética , Embrión de Mamíferos/metabolismo , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Proteínas Nucleares/metabolismo , Transducción de Señal
4.
Cell ; 184(11): 2973-2987.e18, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33945788

RESUMEN

Stony corals are colonial cnidarians that sustain the most biodiverse marine ecosystems on Earth: coral reefs. Despite their ecological importance, little is known about the cell types and molecular pathways that underpin the biology of reef-building corals. Using single-cell RNA sequencing, we define over 40 cell types across the life cycle of Stylophora pistillata. We discover specialized immune cells, and we uncover the developmental gene expression dynamics of calcium-carbonate skeleton formation. By simultaneously measuring the transcriptomes of coral cells and the algae within them, we characterize the metabolic programs involved in symbiosis in both partners. We also trace the evolution of these coral cell specializations by phylogenetic integration of multiple cnidarian cell type atlases. Overall, this study reveals the molecular and cellular basis of stony coral biology.


Asunto(s)
Antozoos/genética , Antozoos/metabolismo , Animales , Antozoos/crecimiento & desarrollo , Biomineralización/genética , Biomineralización/fisiología , Calcinosis/genética , Calcinosis/metabolismo , Arrecifes de Coral , Ecosistema , Inmunidad/genética , Filogenia , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Simbiosis/genética
5.
Cell ; 184(11): 2825-2842.e22, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33932341

RESUMEN

Mouse embryonic development is a canonical model system for studying mammalian cell fate acquisition. Recently, single-cell atlases comprehensively charted embryonic transcriptional landscapes, yet inference of the coordinated dynamics of cells over such atlases remains challenging. Here, we introduce a temporal model for mouse gastrulation, consisting of data from 153 individually sampled embryos spanning 36 h of molecular diversification. Using algorithms and precise timing, we infer differentiation flows and lineage specification dynamics over the embryonic transcriptional manifold. Rapid transcriptional bifurcations characterize the commitment of early specialized node and blood cells. However, for most lineages, we observe combinatorial multi-furcation dynamics rather than hierarchical transcriptional transitions. In the mesoderm, dozens of transcription factors combinatorially regulate multifurcations, as we exemplify using time-matched chimeric embryos of Foxc1/Foxc2 mutants. Our study rejects the notion of differentiation being governed by a series of binary choices, providing an alternative quantitative model for cell fate acquisition.


Asunto(s)
Desarrollo Embrionario/fisiología , Gastrulación/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Embrión de Mamíferos/citología , Desarrollo Embrionario/genética , Femenino , Expresión Génica , Ratones/embriología , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones , Embarazo , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos
6.
Cell ; 176(4): 775-789.e18, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30595452

RESUMEN

Tumor immune cell compositions play a major role in response to immunotherapy, but the heterogeneity and dynamics of immune infiltrates in human cancer lesions remain poorly characterized. Here, we identify conserved intratumoral CD4 and CD8 T cell behaviors in scRNA-seq data from 25 melanoma patients. We discover a large population of CD8 T cells showing continuous progression from an early effector "transitional" into a dysfunctional T cell state. CD8 T cells that express a complete cytotoxic gene set are rare, and TCR sharing data suggest their independence from the transitional and dysfunctional cell states. Notably, we demonstrate that dysfunctional T cells are the major intratumoral proliferating immune cell compartment and that the intensity of the dysfunctional signature is associated with tumor reactivity. Our data demonstrate that CD8 T cells previously defined as exhausted are in fact a highly proliferating, clonal, and dynamically differentiating cell population within the human tumor microenvironment.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Melanoma/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Microambiente Tumoral/inmunología
7.
Cell ; 173(6): 1520-1534.e20, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29856957

RESUMEN

The emergence and diversification of cell types is a leading factor in animal evolution. So far, systematic characterization of the gene regulatory programs associated with cell type specificity was limited to few cell types and few species. Here, we perform whole-organism single-cell transcriptomics to map adult and larval cell types in the cnidarian Nematostella vectensis, a non-bilaterian animal with complex tissue-level body-plan organization. We uncover eight broad cell classes in Nematostella, including neurons, cnidocytes, and digestive cells. Each class comprises different subtypes defined by the expression of multiple specific markers. In particular, we characterize a surprisingly diverse repertoire of neurons, which comparative analysis suggests are the result of lineage-specific diversification. By integrating transcription factor expression, chromatin profiling, and sequence motif analysis, we identify the regulatory codes that underlie Nematostella cell-specific expression. Our study reveals cnidarian cell type complexity and provides insights into the evolution of animal cell-specific genomic regulation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Neuronas/fisiología , ARN , Anémonas de Mar/fisiología , Actinas/química , Secuencias de Aminoácidos , Animales , Cromatina/metabolismo , Análisis por Conglomerados , Perfilación de la Expresión Génica , Genoma , Genómica , Filogenia , Anémonas de Mar/genética , Análisis de Secuencia de ARN , Transcriptoma , Tubulina (Proteína)/química
8.
Cell ; 171(3): 557-572.e24, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-29053968

RESUMEN

Chromosome conformation capture technologies have revealed important insights into genome folding. Yet, how spatial genome architecture is related to gene expression and cell fate remains unclear. We comprehensively mapped 3D chromatin organization during mouse neural differentiation in vitro and in vivo, generating the highest-resolution Hi-C maps available to date. We found that transcription is correlated with chromatin insulation and long-range interactions, but dCas9-mediated activation is insufficient for creating TAD boundaries de novo. Additionally, we discovered long-range contacts between gene bodies of exon-rich, active genes in all cell types. During neural differentiation, contacts between active TADs become less pronounced while inactive TADs interact more strongly. An extensive Polycomb network in stem cells is disrupted, while dynamic interactions between neural transcription factors appear in vivo. Finally, cell type-specific enhancer-promoter contacts are established concomitant to gene expression. This work shows that multiple factors influence the dynamics of chromatin interactions in development.


Asunto(s)
Cromatina/metabolismo , Genoma , Neurogénesis , Animales , Factor de Unión a CCCTC , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos , Exones , Expresión Génica , Redes Reguladoras de Genes , Ratones , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo
9.
Cell ; 167(7): 1883-1896.e15, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984734

RESUMEN

In multicellular organisms, dedicated regulatory circuits control cell type diversity and responses. The crosstalk and redundancies within these circuits and substantial cellular heterogeneity pose a major research challenge. Here, we present CRISP-seq, an integrated method for massively parallel single-cell RNA sequencing (RNA-seq) and clustered regularly interspaced short palindromic repeats (CRISPR)-pooled screens. We show that profiling the genomic perturbation and transcriptome in the same cell enables us to simultaneously elucidate the function of multiple factors and their interactions. We applied CRISP-seq to probe regulatory circuits of innate immunity. By sampling tens of thousands of perturbed cells in vitro and in mice, we identified interactions and redundancies between developmental and signaling-dependent factors. These include opposing effects of Cebpb and Irf8 in regulating the monocyte/macrophage versus dendritic cell lineages and differential functions for Rela and Stat1/2 in monocyte versus dendritic cell responses to pathogens. This study establishes CRISP-seq as a broadly applicable, comprehensive, and unbiased approach for elucidating mammalian regulatory circuits.


Asunto(s)
Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Células Dendríticas/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Monocitos/metabolismo
10.
Cell ; 166(5): 1231-1246.e13, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27545347

RESUMEN

Innate lymphoid cells (ILCs) are critical modulators of mucosal immunity, inflammation, and tissue homeostasis, but their full spectrum of cellular states and regulatory landscapes remains elusive. Here, we combine genome-wide RNA-seq, ChIP-seq, and ATAC-seq to compare the transcriptional and epigenetic identity of small intestinal ILCs, identifying thousands of distinct gene profiles and regulatory elements. Single-cell RNA-seq and flow and mass cytometry analyses reveal compartmentalization of cytokine expression and metabolic activity within the three classical ILC subtypes and highlight transcriptional states beyond the current canonical classification. In addition, using antibiotic intervention and germ-free mice, we characterize the effect of the microbiome on the ILC regulatory landscape and determine the response of ILCs to microbial colonization at the single-cell level. Together, our work characterizes the spectrum of transcriptional identities of small intestinal ILCs and describes how ILCs differentially integrate signals from the microbial microenvironment to generate phenotypic and functional plasticity.


Asunto(s)
Microbioma Gastrointestinal , Inmunidad Innata/genética , Intestinos/inmunología , Intestinos/microbiología , Linfocitos/inmunología , Linfocitos/microbiología , Animales , Secuencia de Bases , Cromatina/metabolismo , Citocinas/inmunología , Epigénesis Genética , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Análisis de la Célula Individual , Transcripción Genética
12.
Cell ; 163(7): 1663-77, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26627738

RESUMEN

Within the bone marrow, stem cells differentiate and give rise to diverse blood cell types and functions. Currently, hematopoietic progenitors are defined using surface markers combined with functional assays that are not directly linked with in vivo differentiation potential or gene regulatory mechanisms. Here, we comprehensively map myeloid progenitor subpopulations by transcriptional sorting of single cells from the bone marrow. We describe multiple progenitor subgroups, showing unexpected transcriptional priming toward seven differentiation fates but no progenitors with a mixed state. Transcriptional differentiation is correlated with combinations of known and previously undefined transcription factors, suggesting that the process is tightly regulated. Histone maps and knockout assays are consistent with early transcriptional priming, while traditional transplantation experiments suggest that in vivo priming may still allow for plasticity given strong perturbations. These data establish a reference model and general framework for studying hematopoiesis at single-cell resolution.


Asunto(s)
Hematopoyesis , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Análisis de la Célula Individual , Transcriptoma , Animales , Trasplante de Médula Ósea , Proteínas Potenciadoras de Unión a CCAAT/genética , Técnicas de Inactivación de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo
13.
Cell ; 148(3): 458-72, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22265598

RESUMEN

Chromosomes are the physical realization of genetic information and thus form the basis for its readout and propagation. Here we present a high-resolution chromosomal contact map derived from a modified genome-wide chromosome conformation capture approach applied to Drosophila embryonic nuclei. The data show that the entire genome is linearly partitioned into well-demarcated physical domains that overlap extensively with active and repressive epigenetic marks. Chromosomal contacts are hierarchically organized between domains. Global modeling of contact density and clustering of domains show that inactive domains are condensed and confined to their chromosomal territories, whereas active domains reach out of the territory to form remote intra- and interchromosomal contacts. Moreover, we systematically identify specific long-range intrachromosomal contacts between Polycomb-repressed domains. Together, these observations allow for quantitative prediction of the Drosophila chromosomal contact map, laying the foundation for detailed studies of chromosome structure and function in a genetically tractable system.


Asunto(s)
Drosophila melanogaster/genética , Genoma de los Insectos , Animales , Núcleo Celular/genética , Cromosomas de Insectos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Modelos Estadísticos , Complejo Represivo Polycomb 1
14.
Cell ; 145(5): 773-86, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21620139

RESUMEN

Mammalian CpG islands are key epigenomic elements that were first characterized experimentally as genomic fractions with low levels of DNA methylation. Currently, CpG islands are defined based on their genomic sequences alone. Here, we develop evolutionary models to show that several distinct evolutionary processes generate and maintain CpG islands. One central evolutionary regime resulting in enriched CpG content is driven by low levels of DNA methylation and consequentially low rates of CpG deamination. Another major force forming CpG islands is biased gene conversion that stabilizes constitutively methylated CpG islands by balancing rapid deamination with CpG fixation. Importantly, evolutionary analysis and population genetics data suggest that selection for high CpG content is not a significant factor contributing to conservation of CpGs in differentially methylated regions. The heterogeneous, but not selective, origins of CpG islands have direct implications for the understanding of DNA methylation patterns in healthy and diseased cells.


Asunto(s)
Islas de CpG , Evolución Molecular , Mamíferos/genética , Animales , Metilación de ADN , Desaminación , Conversión Génica , Humanos , Ratones , Modelos Genéticos , Filogenia
16.
Nature ; 587(7834): 377-386, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32894860

RESUMEN

Here we describe the LifeTime Initiative, which aims to track, understand and target human cells during the onset and progression of complex diseases, and to analyse their response to therapy at single-cell resolution. This mission will be implemented through the development, integration and application of single-cell multi-omics and imaging, artificial intelligence and patient-derived experimental disease models during the progression from health to disease. The analysis of large molecular and clinical datasets will identify molecular mechanisms, create predictive computational models of disease progression, and reveal new drug targets and therapies. The timely detection and interception of disease embedded in an ethical and patient-centred vision will be achieved through interactions across academia, hospitals, patient associations, health data management systems and industry. The application of this strategy to key medical challenges in cancer, neurological and neuropsychiatric disorders, and infectious, chronic inflammatory and cardiovascular diseases at the single-cell level will usher in cell-based interceptive medicine in Europe over the next decade.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Atención a la Salud/métodos , Atención a la Salud/tendencias , Medicina/métodos , Medicina/tendencias , Patología , Análisis de la Célula Individual , Inteligencia Artificial , Atención a la Salud/ética , Atención a la Salud/normas , Diagnóstico Precoz , Educación Médica , Europa (Continente) , Femenino , Salud , Humanos , Legislación Médica , Masculino , Medicina/normas
17.
Genes Dev ; 31(10): 959-972, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28607180

RESUMEN

DNA methylation is a key regulator of embryonic stem cell (ESC) biology, dynamically changing between naïve, primed, and differentiated states. The p53 tumor suppressor is a pivotal guardian of genomic stability, but its contributions to epigenetic regulation and stem cell biology are less explored. We report that, in naïve mouse ESCs (mESCs), p53 restricts the expression of the de novo DNA methyltransferases Dnmt3a and Dnmt3b while up-regulating Tet1 and Tet2, which promote DNA demethylation. The DNA methylation imbalance in p53-deficient (p53-/-) mESCs is the result of augmented overall DNA methylation as well as increased methylation landscape heterogeneity. In differentiating p53-/- mESCs, elevated methylation persists, albeit more mildly. Importantly, concomitant with DNA methylation heterogeneity, p53-/- mESCs display increased cellular heterogeneity both in the "naïve" state and upon induced differentiation. This impact of p53 loss on 5-methylcytosine (5mC) heterogeneity was also evident in human ESCs and mouse embryos in vivo. Hence, p53 helps maintain DNA methylation homeostasis and clonal homogeneity, a function that may contribute to its tumor suppressor activity.


Asunto(s)
Metilación de ADN/genética , Regulación de la Expresión Génica/genética , Heterogeneidad Genética , Homeostasis/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Diferenciación Celular/genética , Células Clonales , ADN (Citosina-5-)-Metiltransferasas/genética , Células Madre Embrionarias , Eliminación de Gen , Humanos , Ratones , Proteínas Proto-Oncogénicas/genética
18.
Trends Genet ; 37(10): 919-932, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34020820

RESUMEN

A fundamental characteristic of animal multicellularity is the spatial coexistence of functionally specialized cell types that are all encoded by a single genome sequence. Cell type transcriptional programs are deployed and maintained by regulatory mechanisms that control the asymmetric, differential access to genomic information in each cell. This genome regulation ultimately results in specific cellular phenotypes. However, the emergence, diversity, and evolutionary dynamics of animal cell types remain almost completely unexplored beyond a few species. Single-cell genomics is emerging as a powerful tool to build comprehensive catalogs of cell types and their associated gene regulatory programs in non-traditional model species. We review the current state of sampling efforts across the animal tree of life and challenges ahead for the comparative study of cell type programs. We also discuss how the phylogenetic integration of cell atlases can lead to the development of models of cell type evolution and a phylogenetic taxonomy of cells.


Asunto(s)
Células/clasificación , Células/metabolismo , Evolución Molecular , Genoma/genética , Genómica , Análisis de la Célula Individual , Animales , Humanos , Especificidad de Órganos
19.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33531344

RESUMEN

Hormones control the major biological functions of stress response, growth, metabolism, and reproduction. In animals, these hormones show pronounced seasonality, with different set-points for different seasons. In humans, the seasonality of these hormones remains unclear, due to a lack of datasets large enough to discern common patterns and cover all hormones. Here, we analyze an Israeli health record on 46 million person-years, including millions of hormone blood tests. We find clear seasonal patterns: The effector hormones peak in winter-spring, whereas most of their upstream regulating pituitary hormones peak only months later, in summer. This delay of months is unexpected because known delays in the hormone circuits last hours. We explain the precise delays and amplitudes by proposing and testing a mechanism for the circannual clock: The gland masses grow with a timescale of months due to trophic effects of the hormones, generating a feedback circuit with a natural frequency of about a year that can entrain to the seasons. Thus, humans may show coordinated seasonal set-points with a winter-spring peak in the growth, stress, metabolism, and reproduction axes.


Asunto(s)
Sistema Endocrino/fisiología , Hormonas/sangre , Registros Médicos/estadística & datos numéricos , Periodicidad , Estaciones del Año , Adaptación Fisiológica , Humanos , Estrés Fisiológico
20.
Nat Chem Biol ; 17(11): 1139-1147, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34504322

RESUMEN

The functional activity and differentiation potential of cells are determined by their interactions with surrounding cells. Approaches that allow unbiased characterization of cell states while at the same time providing spatial information are of major value to assess this environmental influence. However, most current techniques are hampered by a tradeoff between spatial resolution and cell profiling depth. Here, we develop a photocage-based technology that allows isolation and in-depth analysis of live cells from regions of interest in complex ex vivo systems, including primary human tissues. The use of a highly sensitive 4-nitrophenyl(benzofuran) cage coupled to a set of nanobodies allows high-resolution photo-uncaging of different cell types in areas of interest. Single-cell RNA-sequencing of spatially defined CD8+ T cells is used to exemplify the feasibility of identifying location-dependent cell states. The technology described here provides a valuable tool for the analysis of spatially defined cells in diverse biological systems, including clinical samples.


Asunto(s)
Benzofuranos/química , Linfocitos T CD8-positivos/citología , Nitrofenoles/química , Análisis de la Célula Individual , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA