Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
bioRxiv ; 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37873375

RESUMEN

Accurate detection of somatic mutations in single tumor cells is greatly desired as it allows us to quantify the single-cell mutation burden and construct the mutation-based phylogenetic tree. Here we developed scNanoSeq chemistry and profiled 842 single cells from 21 human breast cancer samples. The majority of the mutation-based phylogenetic trees comprise a characteristic stem evolution followed by the clonal sweep. We observed the subtype-dependent lengths in the stem evolution. To explain this phenomenon, we propose that the differences are related to different reprogramming required for different subtypes of breast cancer. Furthermore, we reason that the time that the tumor-initiating cell took to acquire the critical clonal-sweep-initiating mutation by random chance set the time limit for the reprogramming process. We refer to this model as a reprogramming and critical mutation co-timing (RCMC) subtype model. Next, in the sweeping clone, we observed that tumor cells undergo a branched evolution with rapidly decreasing selection. In the most recent clades, effectively neutral evolution has been reached, resulting in a substantially large number of mutational heterogeneities. Integrative analysis with 522-713X ultra-deep bulk whole genome sequencing (WGS) further validated this evolution mode. Mutation-based phylogenetic trees also allow us to identify the early branched cells in a few samples, whose phylogenetic trees support the gradual evolution of copy number variations (CNVs). Overall, the development of scNanoSeq allows us to unveil novel insights into breast cancer evolution.

2.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34155143

RESUMEN

A chromosome 1q21.3 region that is frequently amplified in diverse cancer types encodes phosphatidylinositol (PI)-4 kinase IIIß (PI4KIIIß), a key regulator of secretory vesicle biogenesis and trafficking. Chromosome 1q21.3-amplified lung adenocarcinoma (1q-LUAD) cells rely on PI4KIIIß for Golgi-resident PI-4-phosphate (PI4P) synthesis, prosurvival effector protein secretion, and cell viability. Here, we show that 1q-LUAD cells subjected to prolonged PI4KIIIß antagonist treatment acquire tolerance by activating an miR-218-5p-dependent competing endogenous RNA network that up-regulates PI4KIIα, which provides an alternative source of Golgi-resident PI4P that maintains prosurvival effector protein secretion and cell viability. These findings demonstrate an addiction to Golgi-resident PI4P synthesis in a genetically defined subset of cancers.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Cromosomas Humanos Par 1/genética , Amplificación de Genes , Aparato de Golgi/metabolismo , Fosfatos de Fosfatidilinositol/biosíntesis , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Línea Celular Tumoral , Activación Enzimática , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Fosfatos de Fosfatidilinositol/antagonistas & inhibidores , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/genética
3.
Elife ; 102021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33983114

RESUMEN

ARID1A is one of the most frequently mutated epigenetic regulators in a wide spectrum of cancers. Recent studies have shown that ARID1A deficiency induces global changes in the epigenetic landscape of enhancers and promoters. These broad and complex effects make it challenging to identify the driving mechanisms of ARID1A deficiency in promoting cancer progression. Here, we identified the anti-senescence effect of Arid1a deficiency in the progression of pancreatic intraepithelial neoplasia (PanIN) by profiling the transcriptome of individual PanINs in a mouse model. In a human cell line model, we found that ARID1A deficiency upregulates the expression of aldehyde dehydrogenase 1 family member A1 (ALDH1A1), which plays an essential role in attenuating the senescence induced by oncogenic KRAS through scavenging reactive oxygen species. As a subunit of the SWI/SNF chromatin remodeling complex, our ATAC sequencing data showed that ARID1A deficiency increases the accessibility of the enhancer region of ALDH1A1. This study provides the first evidence that ARID1A deficiency promotes pancreatic tumorigenesis by attenuating KRAS-induced senescence through the upregulation of ALDH1A1 expression.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Senescencia Celular , Proteínas de Unión al ADN/deficiencia , Neoplasias Pancreáticas/patología , Factores de Transcripción/deficiencia , Animales , Carcinogénesis , Línea Celular Tumoral , Transformación Celular Neoplásica , Ensamble y Desensamble de Cromatina , Humanos , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Tamoxifeno/administración & dosificación , Transcriptoma
4.
Cell Rep ; 35(3): 109009, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33882319

RESUMEN

Cancer cells function as primary architects of the tumor microenvironment. However, the molecular features of cancer cells that govern stromal cell phenotypes remain unclear. Here, we show that cancer-associated fibroblast (CAF) heterogeneity is driven by lung adenocarcinoma (LUAD) cells at either end of the epithelial-to-mesenchymal transition (EMT) spectrum. LUAD cells that have high expression of the EMT-activating transcription factor ZEB1 reprogram CAFs through a ZEB1-dependent secretory program and direct CAFs to the tips of invasive projections through a ZEB1-driven CAF repulsion process. The EMT, in turn, sensitizes LUAD cells to pro-metastatic signals from CAFs. Thus, CAFs respond to contextual cues from LUAD cells to promote metastasis.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Fibroblastos Asociados al Cáncer/metabolismo , Células Epiteliales/metabolismo , Neoplasias Renales/genética , Neoplasias Pulmonares/genética , Células Madre Mesenquimatosas/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/secundario , alfa-Globulinas/genética , alfa-Globulinas/metabolismo , Animales , Fibroblastos Asociados al Cáncer/patología , Comunicación Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/secundario , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Ratones Transgénicos , Transducción de Señal , Microambiente Tumoral/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
5.
Matrix Biol ; 91-92: 8-18, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32442601

RESUMEN

Tumor progression is marked by dense collagenous matrix accumulations that dynamically reorganize to accommodate a growing and invasive tumor mass. Cancer-associated fibroblasts (CAFs) play an essential role in matrix remodeling and influence other processes in the tumor microenvironment, including angiogenesis, immunosuppression, and invasion. These findings have spawned efforts to elucidate CAF functionality at the single-cell level. Here, we will discuss how those efforts have impacted our understanding of the ways in which CAFs govern matrix remodeling and the influence of matrix remodeling on the development of an immunosuppressive tumor microenvironment.


Asunto(s)
Fibroblastos Asociados al Cáncer/inmunología , Proteínas de la Matriz Extracelular/inmunología , Matriz Extracelular/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Neovascularización Patológica/inmunología , Fibroblastos Asociados al Cáncer/patología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Citocinas/genética , Citocinas/inmunología , Progresión de la Enfermedad , Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Fibrosis , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunidad Innata , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Transducción de Señal , Análisis de la Célula Individual , Células del Estroma/inmunología , Células del Estroma/patología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
6.
Nat Cell Biol ; 21(9): 1113-1126, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31451770

RESUMEN

Cancer-induced immune responses affect tumour progression and therapeutic response. In multiple murine models and clinical datasets, we identified large variations of neutrophils and macrophages that define 'immune subtypes' of triple-negative breast cancer (TNBC), including neutrophil-enriched (NES) and macrophage-enriched subtypes (MES). Different tumour-intrinsic pathways and mutual regulation between macrophages (or monocytes) and neutrophils contribute to the development of a dichotomous myeloid compartment. MES contains predominantly macrophages that are CCR2-dependent and exhibit variable responses to immune checkpoint blockade (ICB). NES exhibits systemic and local accumulation of immunosuppressive neutrophils (or granulocytic myeloid-derived suppressor cells), is resistant to ICB, and contains a minority of macrophages that seem to be unaffected by CCR2 knockout. A MES-to-NES conversion mediated acquired ICB resistance of initially sensitive MES models. Our results demonstrate diverse myeloid cell frequencies, functionality and potential roles in immunotherapies, and highlight the need to better understand the inter-patient heterogeneity of the myeloid compartment.


Asunto(s)
Inmunoterapia , Células Mieloides/inmunología , Neoplasias de la Mama Triple Negativas/terapia , Microambiente Tumoral/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Granulocitos/inmunología , Inmunoterapia/métodos , Macrófagos/inmunología , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Neoplasias de la Mama Triple Negativas/patología
8.
Nat Commun ; 8: 15045, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28429794

RESUMEN

The majority of breast cancer models for drug discovery are based on orthotopic or subcutaneous tumours. Therapeutic responses of metastases, especially microscopic metastases, are likely to differ from these tumours due to distinct cancer-microenvironment crosstalk in distant organs. Here, to recapitulate such differences, we established an ex vivo bone metastasis model, termed bone-in-culture array or BICA, by fragmenting mouse bones preloaded with breast cancer cells via intra-iliac artery injection. Cancer cells in BICA maintain features of in vivo bone micrometastases regarding the microenvironmental niche, gene expression profile, metastatic growth kinetics and therapeutic responses. Through a proof-of-principle drug screening using BICA, we found that danusertib, an inhibitor of the Aurora kinase family, preferentially inhibits bone micrometastases. In contrast, certain histone methyltransferase inhibitors stimulate metastatic outgrowth of indolent cancer cells, specifically in the bone. Thus, BICA can be used to investigate mechanisms involved in bone colonization and to rapidly test drug efficacies on bone micrometastases.


Asunto(s)
Antineoplásicos/farmacología , Aurora Quinasas/antagonistas & inhibidores , Benzamidas/farmacología , Neoplasias Óseas/tratamiento farmacológico , Huesos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Animales , Aurora Quinasas/genética , Aurora Quinasas/metabolismo , Benzamidas/efectos adversos , Compuestos de Bifenilo , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Huesos/enzimología , Huesos/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Morfolinas , Piridonas/efectos adversos , Técnicas de Cultivo de Tejidos , Microambiente Tumoral
9.
Nature ; 544(7649): 250-254, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28371798

RESUMEN

Blockade of angiogenesis can retard tumour growth, but may also paradoxically increase metastasis. This paradox may be resolved by vessel normalization, which involves increased pericyte coverage, improved tumour vessel perfusion, reduced vascular permeability, and consequently mitigated hypoxia. Although these processes alter tumour progression, their regulation is poorly understood. Here we show that type 1 T helper (TH1) cells play a crucial role in vessel normalization. Bioinformatic analyses revealed that gene expression features related to vessel normalization correlate with immunostimulatory pathways, especially T lymphocyte infiltration or activity. To delineate the causal relationship, we used various mouse models with vessel normalization or T lymphocyte deficiencies. Although disruption of vessel normalization reduced T lymphocyte infiltration as expected, reciprocal depletion or inactivation of CD4+ T lymphocytes decreased vessel normalization, indicating a mutually regulatory loop. In addition, activation of CD4+ T lymphocytes by immune checkpoint blockade increased vessel normalization. TH1 cells that secrete interferon-γ are a major population of cells associated with vessel normalization. Patient-derived xenograft tumours growing in immunodeficient mice exhibited enhanced hypoxia compared to the original tumours in immunocompetent humans, and hypoxia was reduced by adoptive TH1 transfer. Our findings elucidate an unexpected role of TH1 cells in vasculature and immune reprogramming. TH1 cells may be a marker and a determinant of both immune checkpoint blockade and anti-angiogenesis efficacy.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Neovascularización Patológica/inmunología , Neovascularización Fisiológica/inmunología , Neovascularización Fisiológica/fisiología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/trasplante , Permeabilidad Capilar , Hipoxia de la Célula/fisiología , Células Endoteliales/inmunología , Células Endoteliales/fisiología , Femenino , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/patología , Neovascularización Patológica/patología , Pericitos/citología , Pericitos/fisiología , Pronóstico , Células TH1/citología , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/trasplante , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Proc Natl Acad Sci U S A ; 110(52): 21083-8, 2013 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-24324171

RESUMEN

Circulating tumor cells (CTCs) enter peripheral blood from primary tumors and seed metastases. The genome sequencing of CTCs could offer noninvasive prognosis or even diagnosis, but has been hampered by low single-cell genome coverage of scarce CTCs. Here, we report the use of the recently developed multiple annealing and looping-based amplification cycles for whole-genome amplification of single CTCs from lung cancer patients. We observed characteristic cancer-associated single-nucleotide variations and insertions/deletions in exomes of CTCs. These mutations provided information needed for individualized therapy, such as drug resistance and phenotypic transition, but were heterogeneous from cell to cell. In contrast, every CTC from an individual patient, regardless of the cancer subtypes, exhibited reproducible copy number variation (CNV) patterns, similar to those of the metastatic tumor of the same patient. Interestingly, different patients with the same lung cancer adenocarcinoma (ADC) shared similar CNV patterns in their CTCs. Even more interestingly, patients of small-cell lung cancer have CNV patterns distinctly different from those of ADC patients. Our finding suggests that CNVs at certain genomic loci are selected for the metastasis of cancer. The reproducibility of cancer-specific CNVs offers potential for CTC-based cancer diagnostics.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Genoma Humano/genética , Neoplasias Pulmonares/genética , Metástasis de la Neoplasia/genética , Células Neoplásicas Circulantes/química , Secuencia de Bases , Análisis por Conglomerados , Exoma/genética , Biblioteca de Genes , Humanos , Neoplasias Pulmonares/diagnóstico , Datos de Secuencia Molecular , Patología Molecular/métodos , Medicina de Precisión/métodos , Análisis de Secuencia de ADN
11.
Science ; 338(6114): 1622-6, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23258894

RESUMEN

Kindred cells can have different genomes because of dynamic changes in DNA. Single-cell sequencing is needed to characterize these genomic differences but has been hindered by whole-genome amplification bias, resulting in low genome coverage. Here, we report on a new amplification method-multiple annealing and looping-based amplification cycles (MALBAC)-that offers high uniformity across the genome. Sequencing MALBAC-amplified DNA achieves 93% genome coverage ≥1x for a single human cell at 25x mean sequencing depth. We detected digitized copy-number variations (CNVs) of a single cancer cell. By sequencing three kindred cells, we were able to identify individual single-nucleotide variations (SNVs), with no false positives detected. We directly measured the genome-wide mutation rate of a cancer cell line and found that purine-pyrimidine exchanges occurred unusually frequently among the newly acquired SNVs.


Asunto(s)
Neoplasias Colorrectales/genética , Variaciones en el Número de Copia de ADN , ADN de Neoplasias/genética , Técnicas de Amplificación de Ácido Nucleico/métodos , Mutación Puntual , Polimorfismo de Nucleótido Simple , Análisis de la Célula Individual , Adenocarcinoma/genética , Línea Celular Tumoral , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Tasa de Mutación , Análisis de Secuencia de ADN/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA