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
2.
Cell ; 184(9): 2471-2486.e20, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33878291

RESUMEN

Metastasis has been considered as the terminal step of tumor progression. However, recent genomic studies suggest that many metastases are initiated by further spread of other metastases. Nevertheless, the corresponding pre-clinical models are lacking, and underlying mechanisms are elusive. Using several approaches, including parabiosis and an evolving barcode system, we demonstrated that the bone microenvironment facilitates breast and prostate cancer cells to further metastasize and establish multi-organ secondary metastases. We uncovered that this metastasis-promoting effect is driven by epigenetic reprogramming that confers stem cell-like properties on cancer cells disseminated from bone lesions. Furthermore, we discovered that enhanced EZH2 activity mediates the increased stemness and metastasis capacity. The same findings also apply to single cell-derived populations, indicating mechanisms distinct from clonal selection. Taken together, our work revealed an unappreciated role of the bone microenvironment in metastasis evolution and elucidated an epigenomic reprogramming process driving terminal-stage, multi-organ metastases.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Microambiente Tumoral , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Dev Cell ; 56(8): 1100-1117.e9, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33878299

RESUMEN

Estrogen receptor-positive (ER+) breast cancer exhibits a strong bone tropism in metastasis. How the bone microenvironment (BME) impacts ER signaling and endocrine therapy remains poorly understood. Here, we discover that the osteogenic niche transiently and reversibly reduces ER expression and activities specifically in bone micrometastases (BMMs), leading to endocrine resistance. As BMMs progress, the ER reduction and endocrine resistance may partially recover in cancer cells away from the osteogenic niche, creating phenotypic heterogeneity in macrometastases. Using multiple approaches, including an evolving barcoding strategy, we demonstrated that this process is independent of clonal selection, and represents an EZH2-mediated epigenomic reprogramming. EZH2 drives ER+ BMMs toward a basal and stem-like state. EZH2 inhibition reverses endocrine resistance. These data exemplify how epigenomic adaptation to BME promotes phenotypic plasticity of metastatic seeds, fosters intra-metastatic heterogeneity, and alters therapeutic responses. Our study provides insights into the clinical enigma of ER+ metastatic recurrences despite endocrine therapies.


Asunto(s)
Adaptación Fisiológica , Huesos/patología , Neoplasias de la Mama/patología , Receptores de Estrógenos/metabolismo , Microambiente Tumoral , Animales , Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Comunicación Celular , Evolución Clonal , Modelos Animales de Enfermedad , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Uniones Comunicantes/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células MCF-7 , Ratones , Micrometástasis de Neoplasia , Osteogénesis , Transducción de Señal
4.
Oncogene ; 40(5): 997-1011, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33323970

RESUMEN

Estrogen receptor alpha gene (ESR1) mutations occur frequently in ER-positive metastatic breast cancer, and confer clinical resistance to aromatase inhibitors. Expression of the ESR1 Y537S mutation induced an epithelial-mesenchymal transition (EMT) with cells exhibiting enhanced migration and invasion potential in vitro. When small subpopulations of Y537S ESR1 mutant cells were injected along with WT parental cells, tumor growth was enhanced with mutant cells becoming the predominant population in distant metastases. Y537S mutant primary xenograft tumors were resistant to the antiestrogen tamoxifen (Tam) as well as to estradiol (E2) withdrawal. Y537S ESR1 mutant primary tumors metastasized efficiently in the absence of E2; however, Tam treatment significantly inhibited metastasis to distant sites. We identified a nine-gene expression signature, which predicted clinical outcomes of ER-positive breast cancer patients, as well as breast cancer metastasis to the lung. Androgen receptor (AR) protein levels were increased in mutant models, and the AR agonist dihydrotestosterone significantly inhibited estrogen-regulated gene expression, EMT, and distant metastasis in vivo, suggesting that AR may play a role in distant metastatic progression of ESR1 mutant tumors.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Receptor alfa de Estrógeno/genética , Receptores Androgénicos/genética , Tamoxifeno/farmacología , Animales , Inhibidores de la Aromatasa/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Dihidrotestosterona/farmacología , Estradiol/metabolismo , Estrógenos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Mutación/genética , Metástasis de la Neoplasia , Receptores Androgénicos/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nat Cancer ; 1(7): 709-722, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-35122036

RESUMEN

Polyclonal metastases frequently arise from clusters of circulating tumor cells (CTCs). CTC clusters metastasize better than single CTCs, but the underlying molecular mechanisms are poorly understood. Here, we show that polyclonal metastatic seeds exhibit higher resistance to natural killer (NK) cell killing. Using breast cancer models, we observed higher proportions of polyclonal lung metastasis in immunocompetent mice compared with mice lacking NK cells. Depleting NK cells selectively increased monoclonal but not polyclonal metastases, suggesting that CTC clusters are less sensitive to NK-mediated suppression. Transcriptional analyses revealed that clusters have elevated expression of cell-cell adhesion and epithelial genes, which is associated with decreased expression of NK cell activating ligands. Furthermore, perturbing tumor cell epithelial status altered NK ligand expression and sensitivity to NK-mediated killing. Collectively, our findings show that NK cells can determine the fate of CTCs of different epithelial and mesenchymal states, and impact metastatic clonal evolution by favoring polyclonal seeding.


Asunto(s)
Neoplasias Pulmonares , Células Neoplásicas Circulantes , Animales , Recuento de Células , Células Asesinas Naturales , Neoplasias Pulmonares/metabolismo , Ratones , Monitorización Inmunológica
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
7.
Trends Cancer ; 5(2): 95-110, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30755309

RESUMEN

Metastasis to bones is determined by both intrinsic traits of metastatic tumor cells and properties appertaining to the bone microenvironment. Bone marrow niches are critical for all major steps of metastasis, including the seeding of disseminated tumor cells (DTCs) to bone, the survival of DTCs and microscopic metastases under dormancy, and the eventual outgrowth of overt metastases. In this review, we discuss the role of bone marrow niches in bone colonization. The emphasis is on complicated and dynamic nature of cancer cells-niche interaction, which may underpin the long-standing mystery of metastasis dormancy, and represent a therapeutic target for elimination of minimal residue diseases and prevention of life-taking, overt metastases.


Asunto(s)
Neoplasias Óseas/complicaciones , Nicho de Células Madre/genética , Humanos , Metástasis de la Neoplasia
8.
Dev Cell ; 45(6): 663-665, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29920271

RESUMEN

The epithelial-to-mesenchymal transition (EMT) is a key driver of cancer metastasis. In this issue of Developmental Cell, Aiello et al. (2018) demonstrate that an EMT mechanism involving protein internalization impacts cell migration, while Reichert et al. (2018) identify epithelial plasticity as a determinant of metastatic organotropism in pancreatic cancer.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias Pancreáticas , Movimiento Celular , Humanos , Metástasis de la Neoplasia
9.
Cancer Res ; 78(4): 845-852, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29437766

RESUMEN

Circulating tumor cell (CTC) clusters may represent one of the key mechanisms initiating the metastasis process. However, the series of pathophysiologic events by which CTC clusters originate, enter the circulation, and reach the distant sites remain to be identified. The cellular and molecular mechanisms that provide survival advantage for CTC clusters during the transit in the blood stream are also still largely unknown. Understanding the biology of CTC clusters is critical to assess this unified scheme employed by cancer and to device strategies to overcome key pathways responsible for their improved metastatic potential. CTC clusters remain an underdeveloped area of research begging the attention of multidisciplinary cancer research teams. Here, we provide insight on existing preclinical evidence on the potential mechanisms leading to CTC cluster formation and dissemination and on processes that may offer survival advantage. We also offer our perspective on future directions to delineate the role of CTC clusters in metastatic cascade and discuss their clinical significance. Cancer Res; 78(4); 845-52. ©2018 AACR.


Asunto(s)
Células Neoplásicas Circulantes/metabolismo , Humanos , Células Neoplásicas Circulantes/patología
10.
Sci China Life Sci ; 60(10): 1125-1132, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29027156

RESUMEN

Breast cancer cells may disseminate early, before tumor diagnosis. Disseminated tumor cells, or DTCs, reside in the bone marrow, and may persist for years or even decades. Some of these cells may be re-activated to resume aggressive growth, and eventually become overt bone metastases. Recent studies have begun to shed light on this complicated process and revealed multiple steps and intermediate states of colonizing DTCs. However, how cancer-host interactions evolve during this process needs to be further understood. Most of our current knowledge of the bone microenvironment is obtained through studies looking for the hematopoietic stem cell (HSC) niche. Although this long-standing question has not yet been resolved, our search for the HSC niche has resulted in a detailed map of various cell types in the bone marrow. Furthermore, various techniques used to find the HSC niche may also be adapted for finding the cancer cell niche. In this article, we will review the recent progress in both the DTC and HSC areas with a focus on their potential microenvironment niches. We will also discuss how to apply what we have learned from HSC studies to map DTCs in the bone context. We hope to stimulate thoughts and ideas to further elucidate the bone colonization process, and develop potential therapeutic interventions.


Asunto(s)
Células de la Médula Ósea/patología , Médula Ósea/patología , Neoplasias/patología , Nicho de Células Madre , Animales , Movimiento Celular , Células Madre Hematopoyéticas/patología , Humanos , Modelos Biológicos , Metástasis de la Neoplasia
11.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...