Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
EMBO J ; 42(24): e113590, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-38073509

RESUMEN

Cells secrete extracellular vesicles (EVs) and non-vesicular extracellular (nano)particles (NVEPs or ENPs) that may play a role in intercellular communication. Tumor-derived EVs have been proposed to induce immune priming of antigen presenting cells or to be immuno-suppressive agents. We suspect that such disparate functions are due to variable compositions in EV subtypes and ENPs. We aimed to characterize the array of secreted EVs and ENPs of murine tumor cell lines. Unexpectedly, we identified virus-like particles (VLPs) from endogenous murine leukemia virus in preparations of EVs produced by many tumor cells. We established a protocol to separate small EVs from VLPs and ENPs. We compared their protein composition and analyzed their functional interaction with target dendritic cells. ENPs were poorly captured and did not affect dendritic cells. Small EVs specifically induced dendritic cell death. A mixed large/dense EV/VLP preparation was most efficient to induce dendritic cell maturation and antigen presentation. Our results call for systematic re-evaluation of the respective proportions and functions of non-viral EVs and VLPs produced by murine tumors and their contribution to tumor progression.


Asunto(s)
Retrovirus Endógenos , Vesículas Extracelulares , Neoplasias , Animales , Ratones , Vesículas Extracelulares/metabolismo , Línea Celular Tumoral , Diferenciación Celular , Células Dendríticas , Neoplasias/metabolismo
4.
J Extracell Vesicles ; 12(8): e12339, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37548263

RESUMEN

Despite an enormous interest in understanding the bioactivity of extracellular vesicles (EV) in physiology and disease for the development of therapeutic applications, the impact of EV preparation methods remains minimally explored. In this study, we implemented density gradient ultracentrifugation combined with size-exclusion chromatography (DG-SEC), differential ultracentrifugation (dUC) and/or stand-alone SEC (sSEC) to fractionate media conditioned by different cancer cells and/or cancer-associated fibroblasts (CAF). EV-enriched but protein-depleted versus EV-depleted but protein-enriched DG-SEC fractions, and EV-containing dUC and sSEC preparations were quality controlled for particle number, protein concentration, selected protein composition and ultrastructure, characterized for their cytokine content, and dose-dependently evaluated for monocyte-derived dendritic cell (MoDC) maturation by measuring surface marker expression and/or cytokine secretion. EV preparations obtained by DG-SEC from media conditioned by different cancer cell lines or CAF, were depleted from soluble immune suppressive cytokines such as VEGF-A and MCP-1 and potently stimulated MoDC maturation. In contrast, EV-containing dUC or sSEC preparations were not depleted from these soluble cytokines and were unable to mature MoDC. Subsequent processing of dUC EV preparations by SEC dose-dependently restored the immunomodulatory bioactivity. Overall, our results demonstrate that method-dependent off-target enrichment of soluble cytokines has implications for the study of EV immunomodulatory bioactivity and warrants careful consideration.


Asunto(s)
Vesículas Extracelulares , Vesículas Extracelulares/metabolismo , Citocinas/metabolismo , Ultracentrifugación
5.
Proc Natl Acad Sci U S A ; 119(17): e2107394119, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35439048

RESUMEN

Tumor associated macrophages (TAMs), which differentiate from circulating monocytes, are pervasive across human cancers and comprise heterogeneous populations. The contribution of tumor-derived signals to TAM heterogeneity is not well understood. In particular, tumors release both soluble factors and extracellular vesicles (EVs), whose respective impact on TAM precursors may be different. Here, we show that triple negative breast cancer cells (TNBCs) release EVs and soluble molecules promoting monocyte differentiation toward distinct macrophage fates. EVs specifically promoted proinflammatory macrophages bearing an interferon response signature. The combination in TNBC EVs of surface CSF-1 promoting survival and cargoes promoting cGAS/STING or other activation pathways led to differentiation of this particular macrophage subset. Notably, macrophages expressing the EV-induced signature were found among patients' TAMs. Furthermore, higher expression of this signature was associated with T cell infiltration and extended patient survival. Together, this data indicates that TNBC-released CSF-1-bearing EVs promote a tumor immune microenvironment associated with a better prognosis in TNBC patients.


Asunto(s)
Vesículas Extracelulares , Neoplasias de la Mama Triple Negativas , Vesículas Extracelulares/fisiología , Humanos , Macrófagos , Neoplasias de la Mama Triple Negativas/patología
6.
Cell Rep Methods ; 2(1): 100136, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35474866

RESUMEN

Extracellular vesicles (EVs) of various types are released or shed from all cells. EVs carry proteins and contain additional protein and nucleic acid cargo that relates to their biogenesis and cell of origin. EV cargo in liquid biopsies is of widespread interest owing to its ability to provide a retrospective snapshot of cell state at the time of EV release. For the purposes of EV cargo analysis and repertoire profiling, multiplex assays are an essential tool in multiparametric analyte studies but are still being developed for high-parameter EV protein detection. Although bead-based EV multiplex analyses offer EV profiling capabilities with conventional flow cytometers, the utilization of EV multiplex assays has been limited by the lack of software analysis tools for such assays. To facilitate robust EV repertoire studies, we developed multiplex analysis post-acquisition analysis (MPAPASS) open-source software for stitched multiplex analysis, EV database-compatible reporting, and visualization of EV repertoires.


Asunto(s)
Vesículas Extracelulares , Estudios Retrospectivos , Vesículas Extracelulares/metabolismo , Citometría de Flujo/métodos , Programas Informáticos
7.
Toxicol Lett ; 360: 33-43, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35181468

RESUMEN

Despite decreased rates of tobacco smoking in many areas, cigarette smoking remains a major contributor to many health problems. Cigarette smoking can reduce immune system functioning while concurrently increasing inflammation. Dendritic cells in the lung exposed to cigarette smoke become stimulated and go on to activate T-cells. Extracellular vesicles (EVs) are nano-sized particles released by cells. They participate in intercellular communication by transferring functional proteins and nucleic acids to recipient cells and have been implicated in immune responses. For example, they can display MHC-peptide complexes to activate T-cells. In the current study, we sought to understand the role of cigarette smoke extract (CSE) on dendritic cell-derived EVs and their capacity to activate and differentiate T-cells. Primary human dendritic cells (iDCs) were exposed to CSE and EVs were separated and characterized. We exposed autologous primary CD4 + T-cells to iDC-EVs and observed T helper cell populations skewing towards Th1 and Th17 phenotypes. As HIV + individuals are disproportionately likely to be current smokers, we also examined the effects of iDC-EVs on acutely infected T-cells as well as on a cell model of HIV latency (ACH-2). We found that in most cases, iDC-CSE EVs tended to reduce p24 release from the acutely infected primary T-cells, albeit with great variability. We did not observe large effects of iDC-EVs or direct CSE exposure on p24 release from the ACH-2 cell line. Together, these data suggest that iDC-CSE EVs have the capacity to modulate the immune responses, in part by pushing T-cells towards Th1 and Th17 phenotypes.


Asunto(s)
Fumar Cigarrillos , Vesículas Extracelulares , Células Dendríticas , Vesículas Extracelulares/metabolismo , Activación de Linfocitos , Replicación Viral
8.
EMBO J ; 40(8): e105492, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33709510

RESUMEN

Cells release diverse types of extracellular vesicles (EVs), which transfer complex signals to surrounding cells. Specific markers to distinguish different EVs (e.g. exosomes, ectosomes, enveloped viruses like HIV) are still lacking. We have developed a proteomic profiling approach for characterizing EV subtype composition and applied it to human Jurkat T cells. We generated an interactive database to define groups of proteins with similar profiles, suggesting release in similar EVs. Biochemical validation confirmed the presence of preferred partners of commonly used exosome markers in EVs: CD81/ADAM10/ITGB1, and CD63/syntenin. We then compared EVs from control and HIV-1-infected cells. HIV infection altered EV profiles of several cellular proteins, including MOV10 and SPN, which became incorporated into HIV virions, and SERINC3, which was re-routed to non-viral EVs in a Nef-dependent manner. Furthermore, we found that SERINC3 controls the surface composition of EVs. Our workflow provides an unbiased approach for identifying candidate markers and potential regulators of EV subtypes. It can be widely applied to in vitro experimental systems for investigating physiological or pathological modifications of EV release.


Asunto(s)
Vesículas Extracelulares/metabolismo , Infecciones por VIH/metabolismo , Proteoma/metabolismo , Células Cultivadas , Células HEK293 , VIH-1 , Humanos , Células Jurkat , Leucosialina/metabolismo , Glicoproteínas de Membrana/metabolismo , ARN Helicasas/metabolismo
9.
Oncoimmunology ; 9(1): 1715767, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32064174

RESUMEN

Stat3 is constitutively activated in several tumor types and plays an essential role in maintaining their malignant phenotype and immunosupression. To take advantage of the promising antitumor activity of Stat3 targeting, it is vital to understand the mechanism by which Stat3 regulates both cell autonomous and non-autonomous processes. Here, we demonstrated that turning off Stat3 constitutive activation in different cancer cell types induces senescence, thus revealing their Stat3 addiction. Taking advantage of the senescence-associated secretory phenotype (SASP) induced by Stat3 silencing (SASP-siStat3), we designed an immunotherapy. The administration of SASP-siStat3 immunotherapy induced a strong inhibition of triple-negative breast cancer and melanoma growth associated with activation of CD4 + T and NK cells. Combining this immunotherapy with anti-PD-1 antibody resulted in survival improvement in mice bearing melanoma. The characterization of the SASP components revealed that type I IFN-related mediators, triggered by the activation of the cyclic GMP-AMP synthase DNA sensing pathway, are important for its immunosurveillance activity. Overall, our findings provided evidence that administration of SASP-siStat3 or low dose of Stat3-blocking agents would benefit patients with Stat3-addicted tumors to unleash an antitumor immune response and to improve the effectiveness of immune checkpoint inhibitors.


Asunto(s)
Melanoma , Neoplasias de la Mama Triple Negativas , Animales , Senescencia Celular , Humanos , Inmunoterapia , Ratones , Dependencia del Oncogén , Factor de Transcripción STAT3/genética
10.
J Extracell Vesicles ; 10(2): e12050, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33391636

RESUMEN

SARS-CoV-2 entry is mediated by binding of the spike protein (S) to the surface receptor ACE2 and subsequent priming by host TMPRSS2 allowing membrane fusion. Here, we produced extracellular vesicles (EVs) exposing ACE2 and demonstrate that ACE2-EVs are efficient decoys for SARS-CoV-2 S protein-containing lentivirus. Reduction of infectivity positively correlates with the level of ACE2, is much more efficient than with soluble ACE2 and further enhanced by the inclusion of TMPRSS2.


Asunto(s)
Enzima Convertidora de Angiotensina 2/química , COVID-19/prevención & control , COVID-19/virología , Enzima Convertidora de Angiotensina 2/fisiología , Células CACO-2/virología , Línea Celular/virología , Vesículas Extracelulares/metabolismo , Humanos , Lentivirus , Receptores Virales/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus , Internalización del Virus
11.
Int Rev Cell Mol Biol ; 349: 213-249, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31759432

RESUMEN

In addition to direct cell-to-cell contact, dendritic cells (DCs) can regulate the onset of adaptive immunity through the secretion of nano-sized membrane structures, called extracellular vesicles (EVs). This novel mode of communication between cells has added a new layer of complexity to the regulation of immune responses. DCs secrete into their environment different types of EVs containing immunomodulatory molecules that have distinct structural and biochemical properties depending on their intracellular site of origin. Exosomes are generated inside multivesicular bodies and are secreted when these compartments fuse with the plasma membrane, whereas microvesicles are formed and released by budding from the cells' plasma membrane. Once outside the cell of origin, these vesicles can reach target cells through membrane receptor-ligand interactions, modifying their physiological state by the transfer of the EV content or by triggering cell signaling at the cells' surface. Particularly, EVs released by DCs contain major histocompatibility complex (MHC) class I and class II molecules able to activate cognate T cells and promote humoral responses. These activities motivated the use of DC-derived EVs in the treatment of cancer, infectious diseases and autoimmune disorders. The therapeutic potential of these vesicles led to the use of EVs from tumor antigen-loaded DCs in cancer clinical trials, although with limited clinical effects. In this review we will focus on the different EVs released by DCs, their composition and biogenesis, together with their proposed functions as immune regulators.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/inmunología , Vesículas Extracelulares , Animales , Vesículas Extracelulares/inmunología , Humanos
12.
Philos Trans R Soc Lond B Biol Sci ; 373(1737)2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29158309

RESUMEN

In the past decade, cell-to-cell communication mediated by exosomes has attracted growing attention from biomedical scientists and physicians, leading to several recent publications in top-tier journals. Exosomes are generally defined as secreted membrane vesicles, or extracellular vesicles (EVs), corresponding to the intraluminal vesicles of late endosomal compartments, which are secreted upon fusion of multi-vesicular endosomes with the cell's plasma membrane. Cells, however, were shown to release other types of EVs, for instance, by direct budding off their plasma membrane. Some of these EVs share with exosomes major biophysical and biochemical characteristics, such as size, density and membrane orientation, which impose difficulties in their efficient separation. Despite frequent claims in the literature, whether exosomes really display more important patho/physiological functions, or are endowed with higher potential as diagnostic or therapeutic tools than other EVs, is not yet convincingly demonstrated. In this opinion article, we describe reasons for this lack of precision knowledge in the current stage of the EV field, we review recently described approaches to overcome these caveats, and we propose ways to improve our knowledge on the respective functions of distinct EVs, which will be crucial for future development of well-designed EV-based clinical applications.This article is part of the discussion meeting issue 'Extracellular vesicles and the tumour microenvironment'.


Asunto(s)
Vesículas Extracelulares/patología , Vesículas Extracelulares/fisiología , Microambiente Tumoral/fisiología , Comunicación Celular , Exosomas/fisiología , Humanos
13.
EMBO J ; 36(20): 3012-3028, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-28923825

RESUMEN

Exosomes, nano-sized secreted extracellular vesicles (EVs), are actively studied for their diagnostic and therapeutic potential. In particular, exosomes secreted by dendritic cells (DCs) have been shown to carry MHC-peptide complexes allowing efficient activation of T lymphocytes, thus displaying potential as promoters of adaptive immune responses. DCs also secrete other types of EVs of different size, subcellular origin and protein composition, whose immune capacities have not been yet compared to those of exosomes. Here, we show that large EVs (lEVs) released by human DCs are as efficient as small EVs (sEVs), including exosomes, to induce CD4+ T-cell activation in vitro When released by immature DCs, however, lEVs and sEVs differ in their capacity to orient T helper (Th) cell responses, the former favouring secretion of Th2 cytokines, whereas the latter promote Th1 cytokine secretion (IFN-γ). Upon DC maturation, however, these functional differences are abolished, and all EVs become able to induce IFN-γ. Our results highlight the need to comprehensively compare the functionalities of EV subtypes in all patho/physiological systems where exosomes are claimed to perform critical roles.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Vesículas Extracelulares/metabolismo , Activación de Linfocitos , Humanos
14.
J Immunol ; 196(10): 4014-29, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27084100

RESUMEN

In this study, we demonstrate that the unlipidated (U) outer membrane protein (Omp) 19 from Brucella spp. is a competitive inhibitor of human cathepsin L. U-Omp19 inhibits lysosome cathepsins and APC-derived microsome activity in vitro and partially inhibits lysosomal cathepsin L activity within live APCs. Codelivery of U-Omp19 with the Ag can reduce intracellular Ag digestion and increases Ag half-life in dendritic cells (DCs). U-Omp19 retains the Ag in Lamp-2(+) compartments after its internalization and promotes a sustained expression of MHC class I/peptide complexes in the cell surface of DCs. Consequently, U-Omp19 enhances Ag cross-presentation by DCs to CD8(+) T cells. U-Omp19 s.c. delivery induces the recruitment of CD11c(+)CD8α(+) DCs and monocytes to lymph nodes whereas it partially limits in vivo Ag proteolysis inside DCs. Accordingly, this protein is able to induce CD8(+) T cell responses in vivo against codelivered Ag. Antitumor responses were elicited after U-Omp19 coadministration, increasing survival of mice in a murine melanoma challenge model. Collectively, these results indicate that a cysteine protease inhibitor from bacterial origin could be a suitable component of vaccine formulations against tumors.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Brucella/inmunología , Brucelosis/inmunología , Linfocitos T CD8-positivos/fisiología , Vacunas contra el Cáncer/inmunología , Catepsinas/metabolismo , Células Dendríticas/inmunología , Inmunoterapia/métodos , Lipoproteínas/metabolismo , Lisosomas/metabolismo , Melanoma/terapia , Animales , Antígenos de Neoplasias/inmunología , Reactividad Cruzada , Femenino , Activación de Linfocitos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Melanoma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos
15.
Cell ; 164(6): 1226-1232, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26967288

RESUMEN

In multicellular organisms, distant cells can exchange information by sending out signals composed of single molecules or, as increasingly exemplified in the literature, via complex packets stuffed with a selection of proteins, lipids, and nucleic acids, called extracellular vesicles (EVs; also known as exosomes and microvesicles, among other names). This Review covers some of the most striking functions described for EV secretion but also presents the limitations on our knowledge of their physiological roles. While there are initial indications that EV-mediated pathways operate in vivo, the actual nature of the EVs involved in these effects still needs to be clarified. Here, we focus on the context of tumor cells and their microenvironment, but similar results and challenges apply to all patho/physiological systems in which EV-mediated communication is proposed to take place.


Asunto(s)
Vesículas Extracelulares/metabolismo , Neoplasias/metabolismo , Animales , Comunicación Celular , Humanos , Neoplasias/patología , Microambiente Tumoral
16.
Proc Natl Acad Sci U S A ; 113(8): E968-77, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26858453

RESUMEN

Extracellular vesicles (EVs) have become the focus of rising interest because of their numerous functions in physiology and pathology. Cells release heterogeneous vesicles of different sizes and intracellular origins, including small EVs formed inside endosomal compartments (i.e., exosomes) and EVs of various sizes budding from the plasma membrane. Specific markers for the analysis and isolation of different EV populations are missing, imposing important limitations to understanding EV functions. Here, EVs from human dendritic cells were first separated by their sedimentation speed, and then either by their behavior upon upward floatation into iodixanol gradients or by immuno-isolation. Extensive quantitative proteomic analysis allowing comparison of the isolated populations showed that several classically used exosome markers, like major histocompatibility complex, flotillin, and heat-shock 70-kDa proteins, are similarly present in all EVs. We identified proteins specifically enriched in small EVs, and define a set of five protein categories displaying different relative abundance in distinct EV populations. We demonstrate the presence of exosomal and nonexosomal subpopulations within small EVs, and propose their differential separation by immuno-isolation using either CD63, CD81, or CD9. Our work thus provides guidelines to define subtypes of EVs for future functional studies.


Asunto(s)
Antígenos CD/metabolismo , Micropartículas Derivadas de Células/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteómica , Biomarcadores/metabolismo , Humanos
17.
Science ; 349(6253): 1232-6, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26229115

RESUMEN

Infected cells detect viruses through a variety of receptors that initiate cell-intrinsic innate defense responses. Cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS) is a cytosolic sensor for many DNA viruses and HIV-1. In response to cytosolic viral DNA, cGAS synthesizes the second messenger 2'3'-cyclic GMP-AMP (cGAMP), which activates antiviral signaling pathways. We show that in cells producing virus, cGAS-synthesized cGAMP can be packaged in viral particles and extracellular vesicles. Viral particles efficiently delivered cGAMP to target cells. cGAMP transfer by viral particles to dendritic cells activated innate immunity and antiviral defenses. Finally, we show that cell-free murine cytomegalovirus and Modified Vaccinia Ankara virus contained cGAMP. Thus, transfer of cGAMP by viruses may represent a defense mechanism to propagate immune responses to uninfected target cells.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Herpesviridae/inmunología , Inmunidad Innata/inmunología , Muromegalovirus/metabolismo , Nucleótidos Cíclicos/metabolismo , Sistemas de Mensajero Secundario , Virus Vaccinia/metabolismo , Vaccinia/inmunología , Virión/metabolismo , Animales , Chlorocebus aethiops , Citosol/inmunología , Citosol/metabolismo , Citosol/virología , Células Dendríticas/virología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Infecciones por VIH/inmunología , VIH-1/genética , VIH-1/metabolismo , Células HeLa , Humanos , Inmunidad Innata/genética , Ratones , Ratones Endogámicos C57BL , Muromegalovirus/genética , Virus Vaccinia/genética , Células Vero , Virión/genética , Ensamble de Virus
18.
Breast Cancer Res ; 16(6): 491, 2014 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-25479686

RESUMEN

INTRODUCTION: The transcription factor GATA3 is involved in mammary gland development and is crucial for the maintenance of the differentiated status of luminal epithelial cells. The role of GATA3 in breast cancer as a tumor suppressor has been established, although insights into the mechanism of GATA3 expression loss are still required. METHODS: Chromatin immunoprecipitation assays were conducted to study progestin modulation of recruitment of transcription factors to GATA3 promoter. We performed western blot and reverse RT-qPCR experiments to explore progestin regulation of GATA3 protein and mRNA expression respectively. Confocal microscopy and in vitro phosphorylation studies were conducted to examine progestin capacity to induce GATA3 serine phosphorylation in its 308 residue. GATA3 participation in progestin-induced breast cancer growth was addressed in in vitro proliferation and in vivo tumor growth experiments. RESULTS: In this study, we demonstrate that progestin-activated progesterone receptor (PR) reduces GATA3 expression through regulation at the transcriptional and post-translational levels in breast cancer cells. In the former mechanism, the histone methyltransferase enhancer of zeste homolog 2 is co-recruited with activated PR to a putative progesterone response element in the GATA3 proximal promoter, increasing H3K27me3 levels and inducing chromatin compaction, resulting in decreased GATA3 mRNA levels. This transcriptional regulation is coupled with increased GATA3 protein turnover through progestin-induced GATA3 phosphorylation at serine 308 followed by 26S proteasome-mediated degradation. Both molecular mechanisms converge to accomplish decreased GATA3 expression levels in breast cancer cells upon PR activation. In addition, we demonstrated that decreased GATA3 levels are required for progestin-induced upregulation of cyclin A2, which mediates the G1 to S phase transition of the cell cycle and was reported to be associated with poor prognosis in breast cancer. Finally, we showed that downregulation of GATA3 is required for progestin stimulation of both in vitro cell proliferation and in vivo tumor growth. CONCLUSIONS: In the present study, we reveal that progestin-induced PR activation leads to loss of GATA3 expression in breast cancer cells through transcriptional and post-translational regulation. Importantly, we demonstrate that GATA3 downregulation is required for progestin-induced upregulation of cyclin A2 and for progestin-induced in vitro and in vivo breast cancer cell growth.


Asunto(s)
Neoplasias de la Mama/genética , Ciclina A2/genética , Factor de Transcripción GATA3/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Animales/genética , Progestinas/metabolismo , Receptores de Progesterona/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Ciclina A2/metabolismo , Regulación hacia Abajo , Femenino , Factor de Transcripción GATA3/metabolismo , Humanos , Neoplasias Mamarias Animales/metabolismo , Ratones , Fosforilación , Receptores de Estrógenos
19.
Curr Opin Cell Biol ; 29: 116-25, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24959705

RESUMEN

Although observed for several decades, the release of membrane-enclosed vesicles by cells into their surrounding environment has been the subject of increasing interest in the past few years, which led to the creation, in 2012, of a scientific society dedicated to the subject: the International Society for Extracellular Vesicles. Convincing evidence that vesicles allow exchange of complex information fuelled this rise in interest. But it has also become clear that different types of secreted vesicles co-exist, with different intracellular origins and modes of formation, and thus probably different compositions and functions. Exosomes are one sub-type of secreted vesicles. They form inside eukaryotic cells in multivesicular compartments, and are secreted when these compartments fuse with the plasma membrane. Interestingly, different families of molecules have been shown to allow intracellular formation of exosomes and their subsequent secretion, which suggests that even among exosomes different sub-types exist.


Asunto(s)
Exosomas/metabolismo , Animales , Transporte Biológico , Membrana Celular/metabolismo , Humanos
20.
Breast Cancer Res ; 15(6): R118, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24345432

RESUMEN

INTRODUCTION: The role of the progesterone receptor (PR) in breast cancer remains a major clinical challenge. Although PR induces mammary tumor growth, its presence in breast tumors is a marker of good prognosis. We investigated coordinated PR rapid and nonclassical transcriptional effects governing breast cancer growth and endocrine therapy resistance. METHODS: We used breast cancer cell lines expressing wild-type and mutant PRs, cells sensitive and resistant to endocrine therapy, a variety of molecular and cellular biology approaches, in vitro proliferation studies and preclinical models to explore PR regulation of cyclin D1 expression, tumor growth, and response to endocrine therapy. We investigated the clinical significance of activator protein 1 (AP-1) and PR interaction in a cohort of 99 PR-positive breast tumors by an immunofluorescence protocol we developed. The prognostic value of AP-1/PR nuclear colocalization in overall survival (OS) was evaluated using Kaplan-Meier method, and Cox model was used to explore said colocalization as an independent prognostic factor for OS. RESULTS: We demonstrated that at the cyclin D1 promoter and through coordinated rapid and transcriptional effects, progestin induces the assembly of a transcriptional complex among AP-1, Stat3, PR, and ErbB-2 which functions as an enhanceosome to drive breast cancer growth. Our studies in a cohort of human breast tumors identified PR and AP-1 nuclear interaction as a marker of good prognosis and better OS in patients treated with tamoxifen (Tam), an anti-estrogen receptor therapy. Rationale for this finding was provided by our demonstration that Tam inhibits rapid and genomic PR effects, rendering breast cancer cells sensitive to its antiproliferative effects. CONCLUSIONS: We here provided novel insight into the paradox of PR action as well as new tools to identify the subgroup of ER+/PR + patients unlikely to respond to ER-targeted therapies.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Receptor ErbB-2/metabolismo , Receptores de Progesterona/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Núcleo Celular/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Acetato de Medroxiprogesterona/farmacología , Ratones Endogámicos BALB C , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Receptor ErbB-2/genética , Estudios Retrospectivos , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Tamoxifeno/uso terapéutico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...