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













Base de datos
Intervalo de año de publicación
2.
Int J Mol Sci ; 24(18)2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37762615

RESUMEN

Allergic diseases, such as food allergies, asthma, and allergic rhinitis, continue to present a significant challenge for a broad cross-section of the population, despite recent advancements in their treatment and prevention [...].


Asunto(s)
Asma , Hipersensibilidad a los Alimentos , Rinitis Alérgica , Humanos , Prevalencia , Rinitis Alérgica/terapia , Asma/terapia , Asma/epidemiología , Hipersensibilidad a los Alimentos/terapia , Hipersensibilidad a los Alimentos/epidemiología
3.
Methods Mol Biol ; 2666: 299-315, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37166673

RESUMEN

Extracellular vesicles (EV) are small (100-1000 nm) particles that cells release into the extracellular space that have become increasingly famous for their potential in regenerative medicine and for their alterations in diseases such as cancer to promote disease progression, in particular for their potential for intercellular communication. However, studying EV can be challenging due to the broad diversity of both the EV themselves as well as the methods used to study them. This chapter aims to help investigators new to the EV field by describing challenges with studying EV, methods for enriching EV, and a simple EV enrichment protocol using differential ultracentrifugation.


Asunto(s)
Vesículas Extracelulares , Medios de Cultivo Condicionados , Ultracentrifugación/métodos , Espacio Extracelular
4.
Front Allergy ; 4: 1149008, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37034151

RESUMEN

The prevalence of food allergy continues to rise globally, carrying with it substantial safety, economic, and emotional burdens. Although preventative strategies do exist, the heterogeneity of allergy trajectories and clinical phenotypes has made it difficult to identify patients who would benefit from these strategies. Therefore, further studies investigating the molecular mechanisms that differentiate these trajectories are needed. Large-scale omics studies have identified key insights into the molecular mechanisms for many different diseases, however the application of these technologies to uncover the drivers of food allergy development is in its infancy. Here we review the use of omics approaches in food allergy and highlight key gaps in knowledge for applying these technologies for the characterization of food allergy development.

5.
J Allergy Clin Immunol ; 151(3): 595-606, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36872039

RESUMEN

Continuing insight into the molecular mechanisms of atopic disorders has enabled the development of biologics to precisely target these diseases. Food allergy (FA) and eosinophilic gastrointestinal disorders (EGIDs) are driven by similar inflammatory molecular mechanisms and exist along the same atopic disease spectrum. Therefore, many of the same biologics are being investigated to target key drivers of mechanisms shared across the disease states. The enormous potential of biologics for the treatment of FA and EGIDs is highlighted by the significant increases in the number of ongoing clinical trials (more than 30) evaluating their use in these disease states, as well as by the recent US Food and Drug Administration approval of dupilumab for the treatment of eosinophilic esophagitis. Here we discuss past and current research into the use of biologics in FA and EGIDs and their potential role in improving treatment options in the future, with the need to have biologics widely clinically available.


Asunto(s)
Productos Biológicos , Enteritis , Esofagitis Eosinofílica , Hipersensibilidad a los Alimentos , Estados Unidos , Humanos , Niño
6.
Allergy ; 77(10): 2937-2948, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35730331

RESUMEN

The incidence of food allergy (FA) has continued to rise over the last several decades, posing significant burdens on health and quality of life. Significant strides into the advancement of FA diagnosis, prevention, and treatment have been made in recent years. In an effort to lower reliance on resource-intensive food challenges, the field has continued work toward the development of highly sensitive and specific assays capable of high-throughput analysis to assist in the diagnosis FA. In looking toward early infancy as a critical period in the development of allergy or acquisition of tolerance, evidence has increasingly suggested that early intervention via the early introduction of food allergens and maintenance of skin barrier function may decrease the risk of FA. As such, large-scale investigations are underway evaluating infant feeding and the impact of emollient and steroid use in infants with dry skin for the prevention of allergy. On the other end of the spectrum, the past few years have been witness to an explosive increase in clinical trials of novel and innovative therapeutic strategies aimed at the treatment of FA in those whom the disease has already manifested. A milestone in the field, 2020 marked the approval of the first drug, oral peanut allergen, for the indication of peanut allergy. With a foundation of promising data supporting the safety and efficacy of single- and multi-allergen oral immunotherapy, current efforts have turned toward the use of probiotics, biologic agents, and modified allergens to optimize and improve upon existing paradigms. Through these advancements, the field hopes to gain footing in the ongoing battle against FA.


Asunto(s)
Hipersensibilidad a los Alimentos , Hipersensibilidad al Cacahuete , Probióticos , Alérgenos , Arachis , Emolientes , Hipersensibilidad a los Alimentos/diagnóstico , Hipersensibilidad a los Alimentos/prevención & control , Humanos , Lactante , Hipersensibilidad al Cacahuete/prevención & control , Calidad de Vida , Esteroides
7.
Nat Cell Biol ; 24(6): 954-967, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35637408

RESUMEN

Epidemiological studies demonstrate an association between breast cancer (BC) and systemic dysregulation of glucose metabolism. However, how BC influences glucose homeostasis remains unknown. We show that BC-derived extracellular vesicles (EVs) suppress pancreatic insulin secretion to impair glucose homeostasis. EV-encapsulated miR-122 targets PKM in ß-cells to suppress glycolysis and ATP-dependent insulin exocytosis. Mice receiving high-miR-122 EVs or bearing BC tumours exhibit suppressed insulin secretion, enhanced endogenous glucose production, impaired glucose tolerance and fasting hyperglycaemia. These effects contribute to tumour growth and are abolished by inhibiting EV secretion or miR-122, restoring PKM in ß-cells or supplementing insulin. Compared with non-cancer controls, patients with BC have higher levels of circulating EV-encapsulated miR-122 and fasting glucose concentrations but lower fasting insulin; miR-122 levels are positively associated with glucose and negatively associated with insulin. Therefore, EV-mediated impairment of whole-body glycaemic control may contribute to tumour progression and incidence of type 2 diabetes in some patients with BC.


Asunto(s)
Neoplasias de la Mama , Diabetes Mellitus Tipo 2 , Vesículas Extracelulares , MicroARNs , Animales , Neoplasias de la Mama/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Vesículas Extracelulares/metabolismo , Femenino , Glucosa/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Secreción de Insulina , Ratones , MicroARNs/genética , MicroARNs/metabolismo
8.
Nat Cell Biol ; 24(5): 793-804, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35469018

RESUMEN

A decline in skeletal muscle mass and low muscular strength are prognostic factors in advanced human cancers. Here we found that breast cancer suppressed O-linked N-acetylglucosamine (O-GlcNAc) protein modification in muscle through extracellular-vesicle-encapsulated miR-122, which targets O-GlcNAc transferase (OGT). Mechanistically, O-GlcNAcylation of ryanodine receptor 1 (RYR1) competed with NEK10-mediated phosphorylation and increased K48-linked ubiquitination and proteasomal degradation; the miR-122-mediated decrease in OGT resulted in increased RYR1 abundance. We further found that muscular protein O-GlcNAcylation was regulated by hypoxia and lactate through HIF1A-dependent OGT promoter activation and was elevated after exercise. Suppressed O-GlcNAcylation in the setting of cancer, through increasing RYR1, led to higher cytosolic Ca2+ and calpain protease activation, which triggered cleavage of desmin filaments and myofibrillar destruction. This was associated with reduced skeletal muscle mass and contractility in tumour-bearing mice. Our findings link O-GlcNAcylation to muscular protein homoeostasis and contractility and reveal a mechanism of cancer-associated muscle dysregulation.


Asunto(s)
MicroARNs , Neoplasias , Acetilglucosamina/metabolismo , Animales , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , N-Acetilglucosaminiltransferasas/genética , Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
9.
J Mammary Gland Biol Neoplasia ; 23(3): 165-176, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29968174

RESUMEN

Extracellular vesicles (EVs) are secreted by many cell types and are increasingly investigated for their role in human diseases including cancer. Here we focus on the secretion and potential physiological function of non-pathological EVs secreted by polarized normal mammary epithelial cells. Using a transwell system to allow formation of epithelial polarity and EV collection from the apical versus basolateral compartments, we found that impaired secretion of EVs by knockdown of RAB27A or RAB27B suppressed the establishment of mammary epithelial polarity, and that addition of apical but not basolateral EVs suppressed epithelial polarity in a dose-dependent manner. This suggests that apical EV secretion contributes to epithelial polarity, and a possible mechanism is through removal of certain intracellular molecules. In contrast, basolateral but not apical EVs promoted migration of mammary epithelial cells in a motility assay. The protein contents of apical and basolateral EVs from MCF10A and primary human mammary epithelial cells were determined by mass spectrometry proteomic analysis, identifying apical-EV-enriched and basolateral-EV-enriched proteins that may contribute to different physiological functions. Most of these proteins differentially secreted by normal mammary epithelial cells through polarized EV release no longer showed polarized secretion in MCF10A-derived transformed epithelial cells. Our results suggest an essential role of EV secretion in normal mammary epithelial polarization and distinct protein contents and functions in apical versus basolateral EVs secreted by polarized mammary epithelia.


Asunto(s)
Polaridad Celular/fisiología , Células Epiteliales/fisiología , Epitelio/fisiología , Vesículas Extracelulares/fisiología , Glándulas Mamarias Humanas/fisiología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Glándulas Mamarias Humanas/metabolismo , Proteómica/métodos , Proteínas de Unión al GTP rab/metabolismo
10.
Nat Cell Biol ; 20(5): 597-609, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29662176

RESUMEN

Cancer and other cells residing in the same niche engage various modes of interactions to synchronize and buffer the negative effects of environmental changes. Extracellular microRNAs (miRNAs) have recently been implicated in the intercellular crosstalk. Here we show a mechanistic model involving breast-cancer-secreted, extracellular-vesicle-encapsulated miR-105, which is induced by the oncoprotein MYC in cancer cells and, in turn, activates MYC signalling in cancer-associated fibroblasts (CAFs) to induce a metabolic program. This results in the capacity of CAFs to display different metabolic features in response to changes in the metabolic environment. When nutrients are sufficient, miR-105-reprogrammed CAFs enhance glucose and glutamine metabolism to fuel adjacent cancer cells. When nutrient levels are low and metabolic by-products accumulate, these CAFs detoxify metabolic wastes, including lactic acid and ammonium, by converting them into energy-rich metabolites. Thus, the miR-105-mediated metabolic reprogramming of stromal cells contributes to sustained tumour growth by conditioning the shared metabolic environment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Proliferación Celular , Reprogramación Celular , Metabolismo Energético , Exosomas/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células del Estroma/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Exosomas/genética , Exosomas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , Células 3T3 NIH , Comunicación Paracrina , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal , Células del Estroma/patología , Factores de Tiempo , Carga Tumoral , Células Tumorales Cultivadas , Microambiente Tumoral
11.
Clin Cancer Res ; 24(10): 2370-2382, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29500278

RESUMEN

Purpose: Preoperative or neoadjuvant therapy (NT) is increasingly used in patients with locally advanced or inflammatory breast cancer to allow optimal surgery and aim for pathologic response. However, many breast cancers are resistant or relapse after treatment. Here, we investigated conjunctive chemotherapy-triggered events occurring systemically and locally, potentially promoting a cancer stem-like cell (CSC) phenotype and contributing to tumor relapse.Experimental Design: We started by comparing the effect of paired pre- and post-NT patient sera on the CSC properties of breast cancer cells. Using cell lines, patient-derived xenograft models, and primary tumors, we investigated the regulation of CSCs and tumor progression by chemotherapy-induced factors.Results: In human patients and mice, we detected a therapy-induced CSC-stimulatory activity in serum, which was attributed to therapy-associated monocytosis leading to systemic elevation of monocyte chemoattractant proteins (MCP). The post-NT hematopoietic regeneration in the bone marrow highlighted both altered monocyte-macrophage differentiation and biased commitment of stimulated hematopoietic stem cells toward monocytosis. Chemotherapeutic agents also induce monocyte expression of MCPs through a JNK-dependent mechanism. Genetic and pharmacologic inhibitions of the MCP-CCR2 pathway blocked chemotherapy's adverse effect on CSCs. Levels of nuclear Notch and ALDH1 were significantly elevated in primary breast cancers following NT, whereas higher levels of CCR2 in pre-NT tumors were associated with a poor response to NT.Conclusions: Our data establish a mechanism of chemotherapy-induced cancer stemness by linking the cellular events in the bone marrow and tumors, and suggest pharmacologic inhibition of CCR2 as a potential cotreatment during conventional chemotherapy in neoadjuvant and adjuvant settings. Clin Cancer Res; 24(10); 2370-82. ©2018 AACR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Leucocitosis/diagnóstico , Leucocitosis/etiología , Monocitos/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunofenotipificación , Ratones , Células Madre Neoplásicas/patología , Receptores CCR2/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Metastasis Rev ; 35(4): 669-676, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27838868

RESUMEN

It has been recognized that cancer-associated mortality is more often a result of the disrupted physiological functions in multiple organs following metastatic dissemination of cancer cells, rather than the presence and growth of the primary tumor. Despite advances in our understanding of the events leading to cancer initiation, growth, and acquisition of invasive properties, we are still unable to effectively treat metastatic disease. It is now being accepted that the secretion of extracellular vesicles, such as exosomes from cancer cells, has a profound impact on the initiation and propagation of metastatic breast cancer. These cancer-secreted vesicles differ from other means of cellular communication due to their capability of bulk delivery and organotropism. Here, we provide an overview of the role of extracellular vesicles in breast cancer metastasis and discuss key areas that may facilitate our understanding of metastatic breast cancer to guide our efforts towards providing better therapies.


Asunto(s)
Neoplasias de la Mama/patología , Vesículas Extracelulares/patología , Animales , Neoplasias de la Mama/ultraestructura , Femenino , Humanos , Metástasis de la Neoplasia
13.
Clin Cancer Res ; 22(15): 3725-33, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27252414

RESUMEN

A growing body of work has shown that cancer metastasis is not a random spontaneous event; rather, it is the culmination of a cascade of priming steps through which a subpopulation of the tumor cells acquires invasive traits while readying a permissive environment, termed the "premetastatic niche," in which distant metastases can occur. Signals from the primary tumor mobilize and adapt immune cells as well as directly communicating with distant niche cells to induce a broad spectrum of adaptations in target organs, including the induction of angiogenesis, inflammation, extracellular matrix remodeling, and metabolic reprogramming. Together, these interactions facilitate the formation of a premetastatic niche composed of a variable mix of resident and recruited immune cells, endothelial cells, and stromal cells connected through a complex signaling network that we are only beginning to understand. Here, we summarize the latest findings on how cancer induces and guides the formation of this premetastatic niche as well as potential prognostic markers and therapeutic targets that may lead to a better understanding and effective treatment of metastatic disease. Clin Cancer Res; 22(15); 3725-33. ©2016 AACR.


Asunto(s)
Neoplasias/etiología , Neoplasias/patología , Animales , Biomarcadores , Matriz Extracelular/metabolismo , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/terapia , Células del Estroma/metabolismo , Células del Estroma/patología , Microambiente Tumoral
14.
Cell Res ; 26(2): 217-28, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26794868

RESUMEN

MicroRNAs (miRNAs) are critical regulators of gene expression, and exert extensive impacts on development, physiology, and disease of eukaryotes. A high degree of parallelism is found in the molecular basis of miRNA biogenesis and action in plants and animals. Recent studies interestingly suggest a potential cross-kingdom action of plant-derived miRNAs, through dietary intake, in regulating mammalian gene expression. Although the source and scope of plant miRNAs detected in mammalian specimens remain controversial, these initial studies inspired us to determine whether plant miRNAs can be detected in Western human sera and whether these plant miRNAs are able to influence gene expression and cellular processes related to human diseases such as cancer. Here we found that Western donor sera contained the plant miRNA miR159, whose abundance in the serum was inversely correlated with breast cancer incidence and progression in patients. In human sera, miR159 was predominantly detected in the extracellular vesicles, and was resistant to sodium periodate oxidation suggesting the plant-originated 2'-O-methylation on the 3' terminal ribose. In breast cancer cells but not non-cancerous mammary epithelial cells, a synthetic mimic of miR159 was capable of inhibiting proliferation by targeting TCF7 that encodes a Wnt signaling transcription factor, leading to a decrease in MYC protein levels. Oral administration of miR159 mimic significantly suppressed the growth of xenograft breast tumors in mice. These results demonstrate for the first time that a plant miRNA can inhibit cancer growth in mammals.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , MicroARNs/genética , MicroARNs/farmacología , ARN de Planta/genética , ARN de Planta/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Epiteliales/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
15.
Nat Cell Biol ; 17(2): 183-94, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25621950

RESUMEN

Reprogrammed glucose metabolism as a result of increased glycolysis and glucose uptake is a hallmark of cancer. Here we show that cancer cells can suppress glucose uptake by non-tumour cells in the premetastatic niche, by secreting vesicles that carry high levels of the miR-122 microRNA. High miR-122 levels in the circulation have been associated with metastasis in breast cancer patients, and we show that cancer-cell-secreted miR-122 facilitates metastasis by increasing nutrient availability in the premetastatic niche. Mechanistically, cancer-cell-derived miR-122 suppresses glucose uptake by niche cells in vitro and in vivo by downregulating the glycolytic enzyme pyruvate kinase. In vivo inhibition of miR-122 restores glucose uptake in distant organs, including brain and lungs, and decreases the incidence of metastasis. These results demonstrate that, by modifying glucose utilization by recipient premetastatic niche cells, cancer-derived extracellular miR-122 is able to reprogram systemic energy metabolism to facilitate disease progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Glucosa/metabolismo , MicroARNs/metabolismo , Astrocitos/metabolismo , Secuencia de Bases , Neoplasias de la Mama/ultraestructura , Bromodesoxiuridina/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Exosomas/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Luciferasas/metabolismo , Pulmón/patología , MicroARNs/genética , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Piruvato Quinasa/metabolismo
16.
Mol Cancer Res ; 12(11): 1597-609, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25103497

RESUMEN

UNLABELLED: Transforming growth factor beta (TGFß) proteins are multitasking cytokines, in which high levels at tumor sites generally correlate with poor prognosis in human patients with cancer. Previously, it was reported that TGFß downregulates the expression of ataxia telangiectasia-mutated (ATM) and mutS homolog 2 (MSH2) in breast cancer cells through an miRNA-mediated mechanism. In this study, expression of a panel of DNA-repair genes was examined, identifying breast cancer 1, early onset (BRCA1) as a target downregulated by TGFß through the miR181 family. Correlations between the expression levels of TGFß1 and the miR181/BRCA1 axis were observed in primary breast tumor specimens. By downregulating BRCA1, ATM, and MSH2, TGFß orchestrates DNA damage response in certain breast cancer cells to induce a "BRCAness" phenotype, including impaired DNA-repair efficiency and synthetic lethality to the inhibition of poly (ADP-ribose) polymerase (PARP). Xenograft tumors with active TGFß signaling exhibited resistance to the DNA-damaging agent doxorubicin but increased sensitivity to the PARP inhibitor ABT-888. Combination of doxorubicin with ABT-888 significantly improved the treatment efficacy in TGFß-active tumors. Thus, TGFß can induce "BRCAness" in certain breast cancers carrying wild-type BRCA genes and enhance the responsiveness to PARP inhibition, and the molecular mechanism behind this is characterized. IMPLICATIONS: These findings enable better selection of patients with sporadic breast cancer for PARP interventions, which have exhibited beneficial effects in patients carrying BRCA mutations.


Asunto(s)
Proteína BRCA1/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Reparación del ADN/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Factor de Crecimiento Transformador beta/farmacología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Reparación del ADN/efectos de los fármacos , Progresión de la Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Doxorrubicina/farmacología , Femenino , Inestabilidad Genómica/efectos de los fármacos , Humanos , Ratones , MicroARNs , Proteína 2 Homóloga a MutS/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
17.
Sci Rep ; 4: 5750, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-25034888

RESUMEN

Growing evidence links tumor progression with chronic inflammatory processes and dysregulated activity of various immune cells. In this study, we demonstrate that various types of macrophages internalize microvesicles, called exosomes, secreted by breast cancer and non-cancerous cell lines. Although both types of exosomes targeted macrophages, only cancer-derived exosomes stimulated NF-κB activation in macrophages resulting in secretion of pro-inflammatory cytokines such as IL-6, TNFα, GCSF, and CCL2. In vivo mouse experiments confirmed that intravenously injected exosomes are efficiently internalized by macrophages in the lung and brain, which correlated with upregulation of inflammatory cytokines. In mice bearing xenografted human breast cancers, tumor-derived exosomes were internalized by macrophages in axillary lymph nodes thereby triggering expression of IL-6. Genetic ablation of Toll-like receptor 2 (TLR2) or MyD88, a critical signaling adaptor in the NF-κB pathway, completely abolished the effect of tumor-derived exosomes. In contrast, inhibition of TLR4 or endosomal TLRs (TLR3/7/8/9) failed to abrogate NF-κB activation by exosomes. We further found that palmitoylated proteins present on the surface of tumor-secreted exosomes contributed to NF-κB activation. Thus, our results highlight a novel mechanism used by breast cancer cells to induce pro-inflammatory activity of distant macrophages through circulating exosomal vesicles secreted during cancer progression.


Asunto(s)
Exosomas/fisiología , Macrófagos/inmunología , FN-kappa B/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Comunicación Celular , Citocinas/metabolismo , Femenino , Humanos , Inmunomodulación , Células MCF-7 , Macrófagos/metabolismo , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Transducción de Señal
18.
Cancer Cell ; 25(4): 501-15, 2014 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-24735924

RESUMEN

Cancer-secreted microRNAs (miRNAs) are emerging mediators of cancer-host crosstalk. Here we show that miR-105, which is characteristically expressed and secreted by metastatic breast cancer cells, is a potent regulator of migration through targeting the tight junction protein ZO-1. In endothelial monolayers, exosome-mediated transfer of cancer-secreted miR-105 efficiently destroys tight junctions and the integrity of these natural barriers against metastasis. Overexpression of miR-105 in nonmetastatic cancer cells induces metastasis and vascular permeability in distant organs, whereas inhibition of miR-105 in highly metastatic tumors alleviates these effects. miR-105 can be detected in the circulation at the premetastatic stage, and its levels in the blood and tumor are associated with ZO-1 expression and metastatic progression in early-stage breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Endotelio Vascular/patología , MicroARNs/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular , Endotelio Vascular/metabolismo , Femenino , Humanos , MicroARNs/genética , Metástasis de la Neoplasia
19.
Mol Cell Endocrinol ; 382(1): 598-602, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23562748

RESUMEN

There is increasing evidence for the cancer stem cell model in which a subset of cancer cells possessing stem cell properties, referred to as tumor-initiating or cancer stem-like cells (CSCs), play crucial roles in multiple aspects of cancer. Recent studies have started to characterize the crucial role of various cytokines in the tumor microenvironment in regulating the fate of CSCs. In this review, we summarized some of the latest findings on cytokines that drive breast cancer stemness and their mechanisms of action. These cytokines, including IL-6, IL-8, CCL2 and TGF-ß, are frequently elevated in breast tumors and may hold promise as potential therapeutic targets to eradicate CSCs. In combination with conventional chemotherapy and radiotherapy targeting rapidly proliferating cancer cells, intervention of the cancer stemness-driving cytokines may achieve additional benefits for breast cancer patients by suppressing CSC-promoted cancer progression, recurrence, and drug refractoriness.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Citocinas/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Linaje de la Célula , Femenino , Humanos , Modelos Biológicos , Microambiente Tumoral
20.
Methods Mol Biol ; 916: 317-50, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22914951

RESUMEN

Human embryonic stem cell (hESc) lines are likely the in vitro equivalent of the pluripotent epiblast. hESc express high levels of the extracellular matrix (ECM) laminin integrin receptor α6ß1 and consequently can adhere robustly and be propagated in an undifferentiated state on tissue culture plastic coated with the laminin rich basement membrane preparation, Matrigel, even in the absence of supporting fibroblasts. Such cultures represent a critical step in the development of more defined feeder free cultures of hESc; a goal deemed necessary for regenerative medical applications and have been used as the starting point in some differentiation protocols. However, on standard non-deformable tissue culture plastic hESc either fail or inadequately develop the structural/morphological organization of the epiblast in vivo. By contrast, growth of hESc on appropriately defined mechanically deformable polyacrylamide substrates permits recapitulation of many of these in vivo features. These likely herald differences in the precise nature of the integration of signal transduction pathways from soluble morphogens and represent an unexplored variable in hESc (fate) state space. In this chapter we describe how to establish viable hESc colonies on these functionalized polyacrylamide gels. We suggest this strategy as a prospective in vitro model of the genetics, biochemistry, and cell biology of pre- and early-gastrulation stage human embryos and the permissive and instructive roles that cellular and substrate mechanics might play in early embryonic cell fate decisions. Such knowledge should inform regenerative medical applications aimed at enabling or improving the differentiation of specific cell types from embryonic or induced embryonic stem cells.


Asunto(s)
Resinas Acrílicas/química , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Madre Embrionarias/citología , Matriz Extracelular/metabolismo , Estrés Mecánico , Acrilamidas/síntesis química , Acrilamidas/química , Resinas Acrílicas/metabolismo , Calibración , Polaridad Celular , Colágeno/metabolismo , Cristalización , Combinación de Medicamentos , Módulo de Elasticidad , Glutaral/química , Humanos , Laminina/metabolismo , Ligandos , Proteoglicanos/metabolismo , Tripsina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA