Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(22): 5577-5592.e18, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34644529

RESUMEN

Intratumoral heterogeneity is a critical frontier in understanding how the tumor microenvironment (TME) propels malignant progression. Here, we deconvolute the human pancreatic TME through large-scale integration of histology-guided regional multiOMICs with clinical data and patient-derived preclinical models. We discover "subTMEs," histologically definable tissue states anchored in fibroblast plasticity, with regional relationships to tumor immunity, subtypes, differentiation, and treatment response. "Reactive" subTMEs rich in complex but functionally coordinated fibroblast communities were immune hot and inhabited by aggressive tumor cell phenotypes. The matrix-rich "deserted" subTMEs harbored fewer activated fibroblasts and tumor-suppressive features yet were markedly chemoprotective and enriched upon chemotherapy. SubTMEs originated in fibroblast differentiation trajectories, and transitory states were notable both in single-cell transcriptomics and in situ. The intratumoral co-occurrence of subTMEs produced patient-specific phenotypic and computationally predictable heterogeneity tightly linked to malignant biology. Therefore, heterogeneity within the plentiful, notorious pancreatic TME is not random but marks fundamental tissue organizational units.


Asunto(s)
Neoplasias Pancreáticas/patología , Microambiente Tumoral , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Diferenciación Celular , Proliferación Celular , Epitelio/patología , Matriz Extracelular/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Fenotipo , Células del Estroma/patología , Análisis de Supervivencia , Microambiente Tumoral/inmunología
2.
Immunity ; 55(2): 324-340.e8, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35139353

RESUMEN

The aryl hydrocarbon receptor (AhR) is a sensor of products of tryptophan metabolism and a potent modulator of immunity. Here, we examined the impact of AhR in tumor-associated macrophage (TAM) function in pancreatic ductal adenocarcinoma (PDAC). TAMs exhibited high AhR activity and Ahr-deficient macrophages developed an inflammatory phenotype. Deletion of Ahr in myeloid cells or pharmacologic inhibition of AhR reduced PDAC growth, improved efficacy of immune checkpoint blockade, and increased intra-tumoral frequencies of IFNγ+CD8+ T cells. Macrophage tryptophan metabolism was not required for this effect. Rather, macrophage AhR activity was dependent on Lactobacillus metabolization of dietary tryptophan to indoles. Removal of dietary tryptophan reduced TAM AhR activity and promoted intra-tumoral accumulation of TNFα+IFNγ+CD8+ T cells; provision of dietary indoles blocked this effect. In patients with PDAC, high AHR expression associated with rapid disease progression and mortality, as well as with an immune-suppressive TAM phenotype, suggesting conservation of this regulatory axis in human disease.


Asunto(s)
Tolerancia Inmunológica/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Triptófano/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Humanos , Indoles/inmunología , Indoles/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Microbiota/inmunología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Pronóstico , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Triptófano/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/metabolismo
3.
J Pathol ; 261(4): 413-426, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37768107

RESUMEN

Integration and mining of bioimaging data remains a challenge and lags behind the rapidly expanding digital pathology field. We introduce Hourglass, an open-access analytical framework that streamlines biology-driven visualization, interrogation, and statistical assessment of multiparametric datasets. Cognizant of tissue and clinical heterogeneity, Hourglass systematically organizes observations across spatial and global levels and within patient subgroups. Applied to an extensive bioimaging dataset, Hourglass promptly consolidated a breadth of known interleukin-6 (IL-6) functions via its downstream effector STAT3 and uncovered a so-far unknown sexual dimorphism in the IL-6/STAT3-linked intratumoral T-cell response in human pancreatic cancer. As an R package and cross-platform application, Hourglass facilitates knowledge extraction from multi-layered bioimaging datasets for users with or without computational proficiency and provides unique and widely accessible analytical means to harness insights hidden within heterogeneous tissues at the sample and patient level. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Interleucina-6 , Neoplasias Pancreáticas , Humanos , Interleucina-6/genética , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fenotipo , Reino Unido , Factor de Transcripción STAT3/genética
4.
Br J Cancer ; 128(10): 1916-1921, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36927977

RESUMEN

BACKGROUND: Systemic inflammatory scores may aid prognostication and patient selection for trials. We compared five scores in advanced pancreatic adenocarcinoma (PDAC). METHODS: Unresectable/metastatic PDAC patients enrolled in the Comprehensive Molecular Characterisation of Advanced Pancreatic Ductal Adenocarcinoma for Better Treatment Selection trial (NCT02750657) were included. Patients had pre-treatment biopsies for whole genome and RNA sequencing. CD8 immunohistochemistry was available in a subset. The neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio, Prognostic Nutritional Index, Gustave Roussy Immune Score (GRIm-S), and Memorial Sloan Kettering Prognostic Score (MPS) were calculated. Overall survival (OS) was estimated using Kaplan-Meier methods. Associations between inflammatory scores, clinical/genomic characteristics, and OS were analysed. RESULTS: We analysed 263 patients. High-risk NLR, GRIm-S and MPS were poorly prognostic. The GRIm-S had the highest predictive ability: median OS 6.4 vs. 10 months for high risk vs. low-risk (P < 0.001); HR 2.26 (P < 0.001). ECOG ≥ 1, the basal-like subtype, and low-HRDetect were additional poor prognostic factors (P < 0.01). Inflammatory scores did not associate with RNA-based classifiers or homologous recombination repair deficiency genotypes. High-risk MPS (P = 0.04) and GRIm-S (P = 0.02) patients had lower median CD8 + tumour-infiltrating lymphocytes. CONCLUSIONS: Inflammatory scores incorporating NLR have prognostic value in advanced PDAC. Understanding immunophenotypes of poor-risk patients and using these scores in trials will advance the field.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/patología , Pronóstico , Adenocarcinoma/genética , Adenocarcinoma/patología , Linfocitos/patología , Neutrófilos/patología , Estudios Retrospectivos
5.
EMBO J ; 38(14): e100852, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31267556

RESUMEN

Breast cancer prevention is daunting, yet not an unsurmountable goal. Mammary stem and progenitors have been proposed as the cells-of-origin in breast cancer. Here, we present the concept of limiting these breast cancer precursors as a risk reduction approach in high-risk women. A wealth of information now exists for phenotypic and functional characterization of mammary stem and progenitor cells in mouse and human. Recent work has also revealed the hormonal regulation of stem/progenitor dynamics as well as intrinsic lineage distinctions between mammary epithelial populations. Leveraging these insights, molecular marker-guided chemoprevention is an achievable reality.


Asunto(s)
Neoplasias de la Mama/patología , Glándulas Mamarias Humanas/citología , Células Madre/citología , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Ratones , Transducción de Señal , Células Madre/metabolismo , Células Madre/patología
6.
J Proteome Res ; 21(9): 2224-2236, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35981243

RESUMEN

Driven by the lack of targeted therapies, triple-negative breast cancers (TNBCs) have the worst overall survival of all breast cancer subtypes. Considering that cell surface proteins are favorable drug targets and are predominantly glycosylated, glycoproteome profiling has significant potential to facilitate the identification of much-needed drug targets for TNBCs. Here, we performed N-glycoproteomics on six TNBCs and five normal control (NC) cell lines using hydrazide-based enrichment. Quantitative proteomics and integrative data mining led to the discovery of Plexin-B3 (PLXNB3), a previously undescribed TNBC-enriched cell surface protein. Furthermore, siRNA knockdown and CRISPR-Cas9 editing of in vitro and in vivo models show that PLXNB3 is required for TNBC cell line growth, invasion, and migration. Altogether, we provide insights into N-glycoproteome remodeling associated with TNBCs and functional evaluation of an extracted target, which indicate the surface protein PLXNB3 as a potential therapeutic target for TNBCs.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Moléculas de Adhesión Celular , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso , Moléculas de Adhesión de Célula Nerviosa , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
7.
Trends Immunol ; 40(11): 1053-1070, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31645297

RESUMEN

Hematopoietic stem cells (HSCs) self-renew or differentiate into blood cell lineages following extrinsic cues propagated in specialized niches. Support cells and soluble factors in the niche respond to stress and enable progenitor activity. Metalloproteases (MMPs, ADAMs, ADAMTSs) and their inhibitors (TIMPs) control certain physical and biochemical features of the niche by altering protease-dependent bioavailability of local niche factors (e.g., CXCL12, SCF, TGFß, VEGF), matrix turnover, and cellular interactions. With over 40 examples of diverse metalloprotease substrates known to trigger fate-changing decisions, the spatially confined activity of this multi-member protease family is ideally positioned to constitute a higher order control over hematopoiesis. Comprehension of regulated proteolysis in the bone marrow may fuel innovative strategies to harness HSC fate and function.


Asunto(s)
Matriz Extracelular/metabolismo , Células Madre Hematopoyéticas/fisiología , Metaloproteasas/metabolismo , Animales , Diferenciación Celular , Autorrenovación de las Células , Hematopoyesis , Humanos , Proteolisis , Nicho de Células Madre
8.
Eur Radiol ; 32(10): 6712-6722, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36006427

RESUMEN

OBJECTIVES: Transcriptional classifiers (Bailey, Moffitt and Collison) are key prognostic factors of pancreatic ductal adenocarcinoma (PDAC). Among these classifiers, the squamous, basal-like, and quasimesenchymal subtypes overlap and have inferior survival. Currently, only an invasive biopsy can determine these subtypes, possibly resulting in treatment delay. This study aimed to investigate the association between transcriptional subtypes and an externally validated preoperative CT-based radiomic prognostic score (Rad-score). METHODS: We retrospectively evaluated 122 patients who underwent resection for PDAC. All treatment decisions were determined at multidisciplinary tumor boards. Tumor Rad-score values from preoperative CT were dichotomized into high or llow categories. The primary endpoint was the correlation between the transcriptional subtypes and the Rad-score using multivariable linear regression, adjusting for clinical and histopathological variables (i.e., tumor size). Prediction of overall survival (OS) was secondary endpoint. RESULTS: The Bailey transcriptional classifier significantly associated with the Rad-score (coefficient = 0.31, 95% confidence interval [CI]: 0.13-0.44, p = 0.001). Squamous subtype was associated with high Rad-scores while non-squamous subtype was associated with low Rad-scores (adjusted p = 0.03). Squamous subtype and high Rad-score were both prognostic for OS at multivariable analysis with hazard ratios (HR) of 2.79 (95% CI: 1.12-6.92, p = 0.03) and 4.03 (95% CI: 1.42-11.39, p = 0.01), respectively. CONCLUSIONS: In patients with resectable PDAC, an externally validated prognostic radiomic model derived from preoperative CT is associated with the Bailey transcriptional classifier. Higher Rad-scores were correlated with the squamous subtype, while lower Rad-scores were associated with the less lethal subtypes (immunogenic, ADEX, pancreatic progenitor). KEY POINTS: • The transcriptional subtypes of PDAC have been shown to have prognostic importance but they require invasive biopsy to be assessed. • The Rad-score radiomic biomarker, which is obtained non-invasively from preoperative CT, correlates with the Bailey squamous transcriptional subtype and both are negative prognostic biomarkers. • The Rad-score is a promising non-invasive imaging biomarker for personalizing neoadjuvant approaches in patients undergoing resection for PDAC, although additional validation studies are required.


Asunto(s)
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/diagnóstico por imagen , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/cirugía , Humanos , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/cirugía , Pronóstico , Estudios Retrospectivos , Tomografía Computarizada por Rayos X/métodos , Neoplasias Pancreáticas
9.
Immunity ; 36(1): 105-19, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22284418

RESUMEN

Epithelial cells of mucosal tissues provide a barrier against environmental stress, and keratinocytes are key decision makers for immune cell function in the skin. Currently, epithelial signaling networks that instruct barrier immunity remain uncharacterized. Here we have shown that keratinocyte-specific deletion of a disintegrin and metalloproteinase 17 (Adam17) triggers T helper 2 and/or T helper 17 (Th2 and/or Th17) cell-driven atopic dermatitis and myeloproliferative disease. In vivo and in vitro deficiency of ADAM17 dampened Notch signaling, increasing production of the Th2 cell-polarizing cytokine TSLP and myeloid growth factor G-CSF. Ligand-independent Notch activation was identified as a regulator of AP-1 transcriptional activity, with Notch antagonizing c-Fos recruitment to the promoters of Tslp and Csf3 (G-CSF). Further, skin inflammation was rescued and myeloproliferation ameliorated by delivery of active Notch to Adam17(-)(/-) epidermis. Our findings uncover an essential role of ADAM17 in the adult epidermis, demonstrating a gatekeeper function of the ADAM17-Notch-c-Fos triad in barrier immunity.


Asunto(s)
Proteínas ADAM/metabolismo , Citocinas/metabolismo , Epidermis/enzimología , Epidermis/inmunología , Células Precursoras de Granulocitos/citología , Receptores Notch/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Proteína ADAM17 , Animales , Proliferación Celular , Células Epidérmicas , Eliminación de Gen , Humanos , Inflamación , Queratinocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Receptores Notch/inmunología , Transducción de Señal
10.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt A): 1989-2000, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28578911

RESUMEN

Extracellular vesicles (EVs) have emerged as pivotal mediators of intercellular communications in local and distant microenvironments under patho/physiological conditions. EVs contain bioactive materials such as proteins, RNA transcripts, microRNAs and even DNAs, and recent work on their protein profiles has revealed the existence of metalloproteinases including the cell surface-anchored sheddases ADAMs (a disintegrin and metalloproteinases) and soluble ADAMTSs (ADAMs with thrombospondin motifs) as well as cell surface-bound and soluble MMPs (matrix metalloproteinases) from various cell types and body fluids. EV-associated metalloproteinases can alter the make-up of EVs by ectodomain shedding, exert a shedding activity after being taken up by target cells, or directly contribute to degradation of extracellular matrix surrounding cells. In addition, metalloproteinase-loaded EV cargoes sometimes stimulate critical signaling pathways, actively participating in tumor progression. This review focuses on recent findings and knowledge about metalloproteinases in EV biology, and we discuss their potential involvement in human diseases, highlighting the context of tumor cells and their microenvironment. This article is part of a Special Issue entitled: Matrix Metalloproteinases edited by Rafael Fridman.


Asunto(s)
Proteínas ADAMTS/metabolismo , Micropartículas Derivadas de Células/enzimología , Matriz Extracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Proteolisis , Transducción de Señal , Animales , Micropartículas Derivadas de Células/patología , Humanos , Neoplasias/patología , Dominios Proteicos , Microambiente Tumoral
11.
Ecotoxicol Environ Saf ; 142: 544-554, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28482323

RESUMEN

Bioassays of planarian neoplasia highlight the potential of these organisms as useful standards to assess whether environmental toxins such as cadmium promote tumorigenesis. These studies complement other investigations into the exceptional healing and regeneration of planarians - processes that are driven by a population of active stem cells, or neoblasts, which are likely transformed during planarian tumor growth. Our goal was to determine if planarian tumorigenesis assays are amenable to mechanistic studies of cadmium carcinogenesis. To that end we demonstrate, by examining both counts of cell populations by size, and instances of mitosis, that the activity of the stem cell population can be monitored. We also provide evidence that specific biomodulators can affect the potential of planarian neoplastic growth, in that an inhibitor of metalloproteinases effectively blocked the development of the lesions. From these results, we infer that neoblast activity does respond to cadmium-induced tumor growth, and that metalloproteinases are required for the progression of cancer in the planarian.


Asunto(s)
Cadmio/toxicidad , Carcinógenos/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Modelos Biológicos , Planarias/efectos de los fármacos , Animales , Benchmarking , Pruebas de Carcinogenicidad , Transformación Celular Neoplásica/ultraestructura , Cocarcinogénesis , Mitosis/efectos de los fármacos , Planarias/citología , Regeneración/efectos de los fármacos
12.
Development ; 140(7): 1397-401, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23462470

RESUMEN

Progesterone-RankL paracrine signaling has been proposed as a driver of stem cell expansion in the mammary gland, and Elf5 is essential for the differentiation of mammary epithelial progenitor cells. We demonstrate that Elf5 expression is induced by progesterone and that Elf5 and progesterone cooperate to promote alveolar development. The progesterone receptor and Elf5 are expressed in a mutually exclusive pattern, and we identify RankL as the paracrine mediator of the effects of progesterone on Elf5 expression in CD61+ progenitor cells and their consequent differentiation. Blockade of RankL action prevented progesterone-induced side branching and the expansion of Elf5(+) mature luminal cells. These findings describe a mechanism by which steroid hormones can produce the expansion of steroid hormone receptor-negative mammary epithelial cells.


Asunto(s)
Proteínas de Unión al ADN/genética , Glándulas Mamarias Animales/efectos de los fármacos , Progesterona/farmacología , Ligando RANK/farmacología , Células Madre/metabolismo , Factores de Transcripción/genética , Animales , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Transgénicos , Ligando RANK/metabolismo , Ligando RANK/fisiología , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Receptores de Progesterona/fisiología , Células Madre/fisiología , Factores de Transcripción/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
13.
Nature ; 465(7299): 803-7, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20445538

RESUMEN

Reproductive history is the strongest risk factor for breast cancer after age, genetics and breast density. Increased breast cancer risk is entwined with a greater number of ovarian hormone-dependent reproductive cycles, yet the basis for this predisposition is unknown. Mammary stem cells (MaSCs) are located within a specialized niche in the basal epithelial compartment that is under local and systemic regulation. The emerging role of MaSCs in cancer initiation warrants the study of ovarian hormones in MaSC homeostasis. Here we show that the MaSC pool increases 14-fold during maximal progesterone levels at the luteal dioestrus phase of the mouse. Stem-cell-enriched CD49fhi cells amplify at dioestrus, or with exogenous progesterone, demonstrating a key role for progesterone in propelling this expansion. In aged mice, CD49fhi cells display stasis upon cessation of the reproductive cycle. Progesterone drives a series of events where luminal cells probably provide Wnt4 and RANKL signals to basal cells which in turn respond by upregulating their cognate receptors, transcriptional targets and cell cycle markers. Our findings uncover a dynamic role for progesterone in activating adult MaSCs within the mammary stem cell niche during the reproductive cycle, where MaSCs are putative targets for cell transformation events leading to breast cancer.


Asunto(s)
Envejecimiento/fisiología , Glándulas Mamarias Animales/citología , Progesterona/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos , Animales , Recuento de Células , División Celular/efectos de los fármacos , Transformación Celular Neoplásica , Estrógenos/farmacología , Ciclo Estral/sangre , Ciclo Estral/fisiología , Femenino , Homeostasis/efectos de los fármacos , Integrina alfa6/metabolismo , Ratones , Ovariectomía , Comunicación Paracrina/efectos de los fármacos , Progesterona/sangre , Progesterona/metabolismo , Ligando RANK/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Nicho de Células Madre/citología , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/metabolismo , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt4
14.
Nature ; 468(7320): 98-102, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-20881962

RESUMEN

Breast cancer is one of the most common cancers in humans and will on average affect up to one in eight women in their lifetime in the United States and Europe. The Women's Health Initiative and the Million Women Study have shown that hormone replacement therapy is associated with an increased risk of incident and fatal breast cancer. In particular, synthetic progesterone derivatives (progestins) such as medroxyprogesterone acetate (MPA), used in millions of women for hormone replacement therapy and contraceptives, markedly increase the risk of developing breast cancer. Here we show that the in vivo administration of MPA triggers massive induction of the key osteoclast differentiation factor RANKL (receptor activator of NF-κB ligand) in mammary-gland epithelial cells. Genetic inactivation of the RANKL receptor RANK in mammary-gland epithelial cells prevents MPA-induced epithelial proliferation, impairs expansion of the CD49f(hi) stem-cell-enriched population, and sensitizes these cells to DNA-damage-induced cell death. Deletion of RANK from the mammary epithelium results in a markedly decreased incidence and delayed onset of MPA-driven mammary cancer. These data show that the RANKL/RANK system controls the incidence and onset of progestin-driven breast cancer.


Asunto(s)
Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/patología , Progestinas/efectos adversos , Ligando RANK/metabolismo , Animales , Apoptosis/efectos de la radiación , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Daño del ADN , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/efectos de la radiación , Femenino , Rayos gamma , Integrina alfa6/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Acetato de Medroxiprogesterona/administración & dosificación , Acetato de Medroxiprogesterona/efectos adversos , Ratones , FN-kappa B/metabolismo , Osteoclastos/citología , Fosfoproteínas/análisis , Fosfoproteínas/inmunología , Progestinas/administración & dosificación , Ligando RANK/deficiencia , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/deficiencia , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo
15.
Prostate ; 75(16): 1831-43, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26332574

RESUMEN

BACKGROUND: Altered expression and activity of proteases is implicated in inflammation and cancer progression. An important negative regulator of protease activity is TIMP3 (tissue inhibitor of metalloproteinase 3). TIMP3 expression is lacking in many cancers including advanced prostate cancer, and this may facilitate invasion and metastasis by allowing unrestrained protease activity. METHODS: To investigate the role of TIMP3 in prostate cancer progression, we crossed TIMP3-deficient mice (Timp3(-/-)) to mice with prostate-specific deletion of the tumor suppressor Pten (Pten(-/-)), a well-established mouse model of prostate cancer. Tumor growth and progression were compared between Pten(-/-), Timp3(-/-) and control (Pten(-/-), Timp3(+/+)) mice at 16 weeks of age by histopathology and markers of proliferation, vascularity, and tumor invasion. Metalloproteinase activity within the tumors was assessed by gelatin zymography. Inflammatory infiltrates were assessed by immunohistochemistry for macrophages and lymphocytes whereas expression of cytokines and other inflammatory mediators was assessed by quantitative real time PCR and multiplex ELISA. RESULTS: Increased tumor growth, proliferation index, increased microvascular density, and invasion was observed in Pten(-/-), Timp3(-/-) prostate tumors compared to Pten(-/-), Timp3(+/+) tumors. Tumor cell invasion in Pten(-/-), Timp3(-/-) mice was associated with increased expression of matrix metalloprotease (MMP)-9 and activation of MMP-2. There was markedly increased inflammatory cell infiltration into the TIMP3-deficient prostate tumors along with increased expression of monocyte chemoattractant protein-1, cyclooxygenase-2, TNF-α, and interleukin-1ß; all of which are implicated in inflammation and cancer. CONCLUSIONS: This study provides important insights into the role of altered protease activity in promoting prostate cancer invasion and implicates prostate inflammation as an important promoting factor in prostate cancer progression.


Asunto(s)
Invasividad Neoplásica/genética , Próstata/patología , Neoplasias de la Próstata/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Invasividad Neoplásica/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Inhibidor Tisular de Metaloproteinasa-3/genética
16.
J Cell Sci ; 125(Pt 4): 943-55, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22421365

RESUMEN

RANKL (receptor activator of NF-κB ligand) is a crucial cytokine for regulating diverse biological systems such as innate immunity, bone homeostasis and mammary gland differentiation, operating through activation of its cognate receptor RANK. In these normal physiological processes, RANKL signals through paracrine and/or heterotypic mechanisms where its expression and function is tightly controlled. Numerous pathologies involve RANKL deregulation, such as bone loss, inflammatory diseases and cancer, and aberrant RANK expression has been reported in bone cancer. Here, we investigated the significance of RANK in tumor cells with a particular emphasis on homotypic signaling. We selected RANK-positive mouse osteosarcoma and RANK-negative preosteoblastic MC3T3-E1 cells and subjected them to loss- and gain-of-RANK function analyses. By examining a spectrum of tumorigenic properties, we demonstrate that RANK homotypic signaling has a negligible effect on cell proliferation, but promotes cell motility and anchorage-independent growth of osteosarcoma cells and preosteoblasts. By contrast, establishment of RANK signaling in non-tumorigenic mammary epithelial NMuMG cells promotes their proliferation and anchorage-independent growth, but not motility. Furthermore, RANK activation initiates multiple signaling pathways beyond its canonical target, NF-κB. Among these, biochemical inhibition reveals that Erk1/2 is dominant and crucial for the promotion of anchorage-independent survival and invasion of osteoblastic cells, as well as the proliferation of mammary epithelial cells. Thus, RANK signaling functionally contributes to key tumorigenic properties through a cell-autonomous homotypic mechanism. These data also identify the likely inherent differences between epithelial and mesenchymal cell responsiveness to RANK activation.


Asunto(s)
Movimiento Celular , Proliferación Celular , Células Epiteliales/patología , Osteosarcoma/patología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal , Animales , Comunicación Autocrina , Línea Celular Tumoral , Supervivencia Celular , Transformación Celular Neoplásica/patología , Inhibición de Contacto , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glándulas Mamarias Animales/patología , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Invasividad Neoplásica , Osteoblastos/citología , Osteoblastos/metabolismo , Osteosarcoma/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ligando RANK/metabolismo
17.
J Immunol ; 188(6): 2876-83, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22323541

RESUMEN

Lymphocyte infiltration into epithelial tissues and proinflammatory cytokine release are key steps in autoimmune disease. Although cell-autonomous roles of lymphocytes are well studied in autoimmunity, much less is understood about the stromal factors that dictate immune cell function. Tissue inhibitor of metalloproteinases 3 (TIMP3) controls systemic cytokine bioavailability and signaling by inhibiting the ectodomain shedding of cytokines and their receptors. The role of TIMP3 in cytokine biology is emerging; however, its contribution to cellular immunology remains unknown. In this study, we show that TIMP3 produced by the hepatic stroma regulates the basal lymphocyte populations in the liver and prevents autoimmune hepatitis. TIMP3 deficiency in mice led to spontaneous accumulation and activation of hepatic CD4(+), CD8(+), and NKT cells. Treatment with Con A in a model of polyclonal T lymphocyte activation resulted in a greatly enhanced Th1 cytokine response and acute liver failure, which mechanistically depended on TNF signaling. Bone marrow chimeras demonstrated that TIMP3 derived from the stromal rather than hematopoietic compartment provided protection against autoimmunity. Finally, we identified hepatocytes as the major source of Timp3 in a resting liver, whereas significant Timp3 gene transcription was induced by hepatic stellate cells in the inflamed liver. These results uncover metalloproteinase inhibitors as critical stromal factors in regulating cellular immunity during autoimmune hepatitis.


Asunto(s)
Matriz Extracelular/inmunología , Hepatitis Autoinmune/inmunología , Hígado/inmunología , Subgrupos de Linfocitos T/inmunología , Células TH1/inmunología , Inhibidor Tisular de Metaloproteinasa-3/inmunología , Animales , Separación Celular , Citometría de Flujo , Hepatitis Autoinmune/metabolismo , Hepatitis Autoinmune/patología , Immunoblotting , Inmunohistoquímica , Hígado/citología , Hígado/metabolismo , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/metabolismo , Células TH1/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
18.
Cell Rep Methods ; 4(4): 100741, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38569541

RESUMEN

Deep proteomic profiling of rare cell populations has been constrained by sample input requirements. Here, we present DROPPS (droplet-based one-pot preparation for proteomic samples), an accessible low-input platform that generates high-fidelity proteomic profiles of 100-2,500 cells. By applying DROPPS within the mammary epithelium, we elucidated the connection between mitochondrial activity and clonogenicity, identifying CD36 as a marker of progenitor capacity in the basal cell compartment. We anticipate that DROPPS will accelerate biology-driven proteomic research for a multitude of rare cell populations.


Asunto(s)
Biomarcadores , Antígenos CD36 , Glándulas Mamarias Animales , Proteómica , Células Madre , Proteómica/métodos , Antígenos CD36/metabolismo , Animales , Femenino , Células Madre/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Biomarcadores/metabolismo , Biomarcadores/análisis , Epitelio/metabolismo , Ratones , Humanos , Mitocondrias/metabolismo
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167133, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38531482

RESUMEN

The cytosolic dipeptidyl-aminopeptidase 9 (DPP9) cleaves protein N-termini post-proline or -alanine. Our analysis of DPP9 mRNA expression from the TCGA 'breast cancer' data set revealed that low/intermediate DPP9 levels are associated with poor overall survival of breast cancer patients. To unravel the impact of DPP9 on breast cancer development and progression, the transgenic MMTV-PyMT mouse model of metastasizing breast cancer was used. In addition, tissue- and time-controlled genetic deletion of DPP9 by the Cre-loxP recombination system was done. Despite a delay of tumor onset, a higher number of lung metastases were measured in DPP9-deficient mice compared to controls. In human mammary epithelial cells with oncogenic RAS pathway activation, DPP9 deficiency delayed tumorigenic transformation and accelerated TGF-ß1 induced epithelial-to-mesenchymal transition (EMT) of spheroids. For further analysis of the mechanism, primary breast tumor cells were isolated from the MMTV-PyMT model. DPP9 deficiency in these cells caused cancer cell migration and invasion accompanied by EMT. In absence of DPP9, the EMT transcription factor ZEB1 was stabilized due to insufficient degradation by the proteasome. In summary, low expression of DPP9 appears to decelerate mammary tumorigenesis but favors EMT and metastasis, which establishes DPP9 as a novel dynamic regulator of breast cancer initiation and progression.


Asunto(s)
Neoplasias de la Mama , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Transición Epitelial-Mesenquimal , Animales , Humanos , Femenino , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Ratones , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/metabolismo , Metástasis de la Neoplasia , Regulación Neoplásica de la Expresión Génica , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Línea Celular Tumoral , Ratones Noqueados , Ratones Transgénicos
20.
Proteomics ; 13(10-11): 1624-36, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23526769

RESUMEN

Exosomes are small microvesicles secreted from the late endosomal compartment of cells. Although an increasing body of evidence indicates that they play a pivotal role in cell-to-cell communication, the biological functions of exosomes are far from fully understood. Recent work has revealed detailed proteomic profiles of exosomes from cell lines and body fluids, which may provide clues to understanding their biological significance and general importance in human diseases. Metalloproteinases include the cell surface-anchored sheddases a disintegrin and metalloproteinases, as well as cell surface-bound and soluble matrix metalloproteinases and these extracellular proteases have been detected in exosomes by proteomic analyses. Exosomes play a key role in the transfer of proteins to other cells and metalloproteinases may provide a novel platform where ectodomain shedding by these membrane proteases alters the makeup of the recipient cell's surface. This review aims to address some of the facets of exosome biology with particular emphasis on the proteolytic factors and we discuss their potential involvement in human diseases, especially tumor biology.


Asunto(s)
Exosomas/enzimología , Metaloproteasas/metabolismo , Animales , Exosomas/metabolismo , Humanos , Transporte de Proteínas , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA