Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 5(1): 1263, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36400937

RESUMEN

Upcoming technologies enable routine collection of highly multiplexed (20-60 channel), subcellular resolution images of mammalian tissues for research and diagnosis. Extracting single cell data from such images requires accurate image segmentation, a challenging problem commonly tackled with deep learning. In this paper, we report two findings that substantially improve image segmentation of tissues using a range of machine learning architectures. First, we unexpectedly find that the inclusion of intentionally defocused and saturated images in training data substantially improves subsequent image segmentation. Such real augmentation outperforms computational augmentation (Gaussian blurring). In addition, we find that it is practical to image the nuclear envelope in multiple tissues using an antibody cocktail thereby better identifying nuclear outlines and improving segmentation. The two approaches cumulatively and substantially improve segmentation on a wide range of tissue types. We speculate that the use of real augmentations will have applications in image processing outside of microscopy.


Asunto(s)
Aprendizaje Profundo , Humanos , Animales , Procesamiento de Imagen Asistido por Computador/métodos , Aprendizaje Automático , Núcleo Celular , Mamíferos
2.
Cell Genom ; 2(8)2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-36419822

RESUMEN

Intra-tumor heterogeneity (ITH) of human tumors is important for tumor progression, treatment response, and drug resistance. However, the spatial distribution of ITH remains incompletely understood. Here, we present spatial analysis of ITH in lung adenocarcinomas from 147 patients using multi-region mass spectrometry of >5,000 regions, single-cell copy number sequencing of ~2,000 single cells, and cyclic immunofluorescence of >10 million cells. We identified two distinct spatial patterns among tumors, termed clustered and random geographic diversification (GD). These patterns were observed in the same samples using both proteomic and genomic data. The random proteomic GD pattern, which is characterized by decreased cell adhesion and lower levels of tumor-interacting endothelial cells, was significantly associated with increased risk of recurrence or death in two independent patient cohorts. Our study presents comprehensive spatial mapping of ITH in lung adenocarcinoma and provides insights into the mechanisms and clinical consequences of GD.

3.
Science ; 376(6592): eabi8175, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35482859

RESUMEN

Establishing causal relationships between genetic alterations of human cancers and specific phenotypes of malignancy remains a challenge. We sequentially introduced mutations into healthy human melanocytes in up to five genes spanning six commonly disrupted melanoma pathways, forming nine genetically distinct cellular models of melanoma. We connected mutant melanocyte genotypes to malignant cell expression programs in vitro and in vivo, replicative immortality, malignancy, rapid tumor growth, pigmentation, metastasis, and histopathology. Mutations in malignant cells also affected tumor microenvironment composition and cell states. Our melanoma models shared genotype-associated expression programs with patient melanomas, and a deep learning model showed that these models partially recapitulated genotype-associated histopathological features as well. Thus, a progressive series of genome-edited human cancer models can causally connect genotypes carrying multiple mutations to phenotype.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Melanocitos/metabolismo , Melanoma/patología , Mutación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Microambiente Tumoral/genética
4.
Cancer Discov ; 12(6): 1518-1541, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35404441

RESUMEN

Cutaneous melanoma is a highly immunogenic malignancy that is surgically curable at early stages but life-threatening when metastatic. Here we integrate high-plex imaging, 3D high-resolution microscopy, and spatially resolved microregion transcriptomics to study immune evasion and immunoediting in primary melanoma. We find that recurrent cellular neighborhoods involving tumor, immune, and stromal cells change significantly along a progression axis involving precursor states, melanoma in situ, and invasive tumor. Hallmarks of immunosuppression are already detectable in precursor regions. When tumors become locally invasive, a consolidated and spatially restricted suppressive environment forms along the tumor-stromal boundary. This environment is established by cytokine gradients that promote expression of MHC-II and IDO1, and by PD1-PDL1-mediated cell contacts involving macrophages, dendritic cells, and T cells. A few millimeters away, cytotoxic T cells synapse with melanoma cells in fields of tumor regression. Thus, invasion and immunoediting can coexist within a few millimeters of each other in a single specimen. SIGNIFICANCE: The reorganization of the tumor ecosystem in primary melanoma is an excellent setting in which to study immunoediting and immune evasion. Guided by classic histopathology, spatial profiling of proteins and mRNA reveals recurrent morphologic and molecular features of tumor evolution that involve localized paracrine cytokine signaling and direct cell-cell contact. This article is highlighted in the In This Issue feature, p. 1397.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Citocinas , Ecosistema , Humanos , Melanoma/patología , Neoplasias Cutáneas/genética , Melanoma Cutáneo Maligno
5.
Nat Methods ; 19(3): 311-315, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34824477

RESUMEN

Highly multiplexed tissue imaging makes detailed molecular analysis of single cells possible in a preserved spatial context. However, reproducible analysis of large multichannel images poses a substantial computational challenge. Here, we describe a modular and open-source computational pipeline, MCMICRO, for performing the sequential steps needed to transform whole-slide images into single-cell data. We demonstrate the use of MCMICRO on tissue and tumor images acquired using multiple imaging platforms, thereby providing a solid foundation for the continued development of tissue imaging software.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Neoplasias , Diagnóstico por Imagen , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Programas Informáticos
6.
Cancer Cell ; 39(12): 1623-1642.e20, 2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34739845

RESUMEN

The mechanisms regulating exhaustion of tumor-infiltrating lymphocytes (TIL) and responsiveness to PD-1 blockade remain partly unknown. In human ovarian cancer, we show that tumor-specific CD8+ TIL accumulate in tumor islets, where they engage antigen and upregulate PD-1, which restrains their functions. Intraepithelial PD-1+CD8+ TIL can be, however, polyfunctional. PD-1+ TIL indeed exhibit a continuum of exhaustion states, with variable levels of CD28 costimulation, which is provided by antigen-presenting cells (APC) in intraepithelial tumor myeloid niches. CD28 costimulation is associated with improved effector fitness of exhausted CD8+ TIL and is required for their activation upon PD-1 blockade, which also requires tumor myeloid APC. Exhausted TIL lacking proper CD28 costimulation in situ fail to respond to PD-1 blockade, and their response may be rescued by local CTLA-4 blockade and tumor APC stimulation via CD40L.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Antígenos CD28/metabolismo , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Células Mieloides/metabolismo , Neoplasias/tratamiento farmacológico , Nicho de Células Madre/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias/inmunología
7.
Cell ; 183(7): 1848-1866.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33301708

RESUMEN

Obesity is a major cancer risk factor, but how differences in systemic metabolism change the tumor microenvironment (TME) and impact anti-tumor immunity is not understood. Here, we demonstrate that high-fat diet (HFD)-induced obesity impairs CD8+ T cell function in the murine TME, accelerating tumor growth. We generate a single-cell resolution atlas of cellular metabolism in the TME, detailing how it changes with diet-induced obesity. We find that tumor and CD8+ T cells display distinct metabolic adaptations to obesity. Tumor cells increase fat uptake with HFD, whereas tumor-infiltrating CD8+ T cells do not. These differential adaptations lead to altered fatty acid partitioning in HFD tumors, impairing CD8+ T cell infiltration and function. Blocking metabolic reprogramming by tumor cells in obese mice improves anti-tumor immunity. Analysis of human cancers reveals similar transcriptional changes in CD8+ T cell markers, suggesting interventions that exploit metabolism to improve cancer immunotherapy.


Asunto(s)
Inmunidad , Neoplasias/inmunología , Neoplasias/metabolismo , Obesidad/metabolismo , Microambiente Tumoral , Adiposidad , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Células HEK293 , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Cinética , Linfocitos Infiltrantes de Tumor , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Análisis de Componente Principal , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteómica
8.
Cell ; 183(5): 1354-1366.e13, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33065030

RESUMEN

The COVID-19 pandemic has led to extensive morbidity and mortality throughout the world. Clinical features that drive SARS-CoV-2 pathogenesis in humans include inflammation and thrombosis, but the mechanistic details underlying these processes remain to be determined. In this study, we demonstrate endothelial disruption and vascular thrombosis in histopathologic sections of lungs from both humans and rhesus macaques infected with SARS-CoV-2. To define key molecular pathways associated with SARS-CoV-2 pathogenesis in macaques, we performed transcriptomic analyses of bronchoalveolar lavage and peripheral blood and proteomic analyses of serum. We observed macrophage infiltrates in lung and upregulation of macrophage, complement, platelet activation, thrombosis, and proinflammatory markers, including C-reactive protein, MX1, IL-6, IL-1, IL-8, TNFα, and NF-κB. These results suggest a model in which critical interactions between inflammatory and thrombosis pathways lead to SARS-CoV-2-induced vascular disease. Our findings suggest potential therapeutic targets for COVID-19.


Asunto(s)
COVID-19/complicaciones , COVID-19/inmunología , SARS-CoV-2/genética , Trombosis/complicaciones , Enfermedades Vasculares/complicaciones , Anciano de 80 o más Años , Animales , Lavado Broncoalveolar , Proteína C-Reactiva/análisis , COVID-19/sangre , COVID-19/patología , Activación de Complemento , Citocinas/sangre , Femenino , Humanos , Inflamación/sangre , Inflamación/inmunología , Inflamación/virología , Pulmón/patología , Macaca mulatta , Macrófagos/inmunología , Masculino , Activación Plaquetaria , Trombosis/sangre , Trombosis/patología , Transcriptoma , Enfermedades Vasculares/sangre , Enfermedades Vasculares/patología
10.
Nat Commun ; 11(1): 1459, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32193378

RESUMEN

Combined PARP and immune checkpoint inhibition has yielded encouraging results in ovarian cancer, but predictive biomarkers are lacking. We performed immunogenomic profiling and highly multiplexed single-cell imaging on tumor samples from patients enrolled in a Phase I/II trial of niraparib and pembrolizumab in ovarian cancer (NCT02657889). We identify two determinants of response; mutational signature 3 reflecting defective homologous recombination DNA repair, and positive immune score as a surrogate of interferon-primed exhausted CD8 + T-cells in the tumor microenvironment. Presence of one or both features associates with an improved outcome while concurrent absence yields no responses. Single-cell spatial analysis reveals prominent interactions of exhausted CD8 + T-cells and PD-L1 + macrophages and PD-L1 + tumor cells as mechanistic determinants of response. Furthermore, spatial analysis of two extreme responders shows differential clustering of exhausted CD8 + T-cells with PD-L1 + macrophages in the first, and exhausted CD8 + T-cells with cancer cells harboring genomic PD-L1 and PD-L2 amplification in the second.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno B7-H1/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Análisis Mutacional de ADN , Monitoreo de Drogas/métodos , Femenino , Amplificación de Genes , Humanos , Indazoles/farmacología , Indazoles/uso terapéutico , Interferones/inmunología , Interferones/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Ovario/patología , Piperidinas/farmacología , Piperidinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Reparación del ADN por Recombinación/genética , Análisis de la Célula Individual , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
11.
Cell Syst ; 8(5): 412-426.e7, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31078528

RESUMEN

Tyrosine kinase inhibitors (TKIs) are widely used to treat solid tumors but can be cardiotoxic. The molecular basis for this toxicity and its relationship to therapeutic mechanisms remain unclear; we therefore undertook a systems-level analysis of human cardiomyocytes (CMs) exposed to four TKIs. CMs differentiated from human induced pluripotent stem cells (hiPSCs) were exposed to sunitinib, sorafenib, lapatinib, or erlotinib, and responses were assessed by functional assays, microscopy, RNA sequencing, and mass spectrometry (GEO: GSE114686; PRIDE: PXD012043). TKIs have diverse effects on hiPSC-CMs distinct from inhibition of tyrosine-kinase-mediated signal transduction; cardiac metabolism is particularly sensitive. Following sorafenib treatment, oxidative phosphorylation is downregulated, resulting in a profound defect in mitochondrial energetics. Cells adapt by upregulating aerobic glycolysis. Adaptation makes cells less acutely sensitive to sorafenib but may have long-term negative consequences. Thus, CMs exhibit adaptive responses to anti-cancer drugs conceptually similar to those previously shown in tumors to mediate drug resistance.


Asunto(s)
Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Aclimatación , Antineoplásicos/farmacología , Cardiotoxicidad/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Clorhidrato de Erlotinib/farmacología , Perfilación de la Expresión Génica/métodos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lapatinib/farmacología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Sorafenib/farmacología , Sunitinib/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...