Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Adv Drug Deliv Rev ; 176: 113838, 2021 09.
Article in English | MEDLINE | ID: mdl-34144088

ABSTRACT

Restoring effective anti-tumor immune responses to cure cancer is a promising strategy, but challenging to achieve due to the intricate crosstalk between tumor and immune cells. While it is established that tumor cells acquire traits to escape immune recognition, the involvement of extracellular vesicles (EVs) in curbing immune cell activation is rapidly emerging. By assisting cancer cells in spreading immunomodulatory signals in the form of (glyco)proteins, lipids, nucleic acids and metabolic regulators, EVs recently emerged as versatile mediators of immune suppression. Blocking their action might reactivate immune cell function and natural antitumor immune responses. Alternatively, EV communication may be exploited to boost anti-tumor immunity. Indeed, novel insights into EV biology paved the way for efficient ex vivo production of 'rationally engineered' EVs that function as potent antitumor vaccines or carry out specific functional tasks. In this review we discuss the latest findings on immune regulation by cancer EVs and explore how EV-mediated communication can be either targeted or harnessed to restore immunity as a means for cancer therapy.


Subject(s)
Extracellular Vesicles/immunology , Immunotherapy/methods , Neoplasms/therapy , Animals , Glycoproteins/immunology , Humans , Lipids/immunology , Neoplasms/immunology , Nucleic Acids/immunology , Signal Transduction/immunology
3.
Pharmaceutics ; 12(10)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081417

ABSTRACT

Extracellular vesicles (EVs) are sophisticated and sensitive messengers released by cells to communicate with and influence distant and neighboring cells via selective transfer of bioactive content, including protein lipids and nucleic acids. EVs have therefore attracted broad interest as new and refined potential therapeutic systems in many diseases, including cancer, due to their low immunogenicity, non-toxicity, and elevated bioavailability. They might serve as safe and effective vehicles for the transport of therapeutic molecules to specific tissues and cells. In this review, we focus on EVs as a vehicle for gene therapy in cancer. We describe recent developments in EV engineering to achieve efficient intracellular delivery of cancer therapeutics and avoid off-target effects, to provide an overview of the potential applications of EV-mediated gene therapy and the most promising biomedical advances.

5.
Crit Rev Oncol Hematol ; 139: 125-127, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30890387

ABSTRACT

Exosome-mediated transfer of regulatory RNAs is a key feature that enables cancer cells to shape a tumor-promoting environment. Cancers growing in the bone can use this communication modality to disrupt the homeostatic balance between bone forming and bone resorbing cells, which results in the release of bone-embedded factors supporting cancer growth and progression. Long noncoding RNAs (lncRNAs) are potent regulators of cell fate determination with exceptional cell- and tissue-specificity that are secreted by cancer cells via exosomes. In multiple myeloma (MM), the exosomal transfer of the lncRNA RUNX2-AS1 specifically inhibits the osteogenic differentiation capacity of mesenchymal stem cells (MSC) by repressing the master regulator of bone formation RUNX2. Detailed studies into the role of exosomal lncRNA transfer in the bone microenvironment in vivo might constitute the basis for the development of novel therapeutic strategies for tumor-associated bone lesions.


Subject(s)
Bone Neoplasms/pathology , Exosomes/metabolism , Mesenchymal Stem Cells/pathology , RNA, Long Noncoding/genetics , Tumor Microenvironment , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Exosomes/genetics , Humans , Mesenchymal Stem Cells/metabolism
6.
J Vis Exp ; (135)2018 05 06.
Article in English | MEDLINE | ID: mdl-29782011

ABSTRACT

Within the tumor microenvironment, resident or recruited mesenchymal stem cells (MSCs) contribute to malignant progression in multiple cancer types. Under the influence of specific environmental signals, these adult stem cells can release paracrine mediators leading to accelerated tumor growth and metastasis. Defining the crosstalk between tumor and MSCs is of primary importance to understand the mechanisms underlying cancer progression and identify novel targets for therapeutic intervention. Cancer cells produce high amounts of extracellular vesicles (EVs), which can profoundly affect the behavior of target cells in the tumor microenvironment or at distant sites. Tumor EVs enclose functional biomolecules, including inflammatory RNAs and (onco)proteins, that can educate stromal cells to enhance the metastatic behavior of cancer cells or to participate in the pre-metastatic niche formation. In this article, we describe the development of a preclinical cancer mouse model that enables specific evaluation of the EV-mediated crosstalk between tumor and mesenchymal stem cells. First, we describe the purification and characterization of tumor-secreted EVs and the assessment of the EV internalization by MSCs. We then make use of a multiplex bead-based immunoassay to evaluate the alteration of the MSC cytokine expression profile induced by cancer EVs. Finally, we illustrate the generation of a bioluminescent orthotopic xenograft mouse model of osteosarcoma that recapitulates the tumor-MSC interaction, and show the contribution of EV-educated MSCs to tumor growth and metastasis formation. Our model provides the opportunity to define how cancer EVs shape a tumor-supporting environment, and to evaluate whether blockade of the EV-mediated communication between tumor and MSCs prevents cancer progression.


Subject(s)
Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Osteosarcoma/diagnosis , Animals , Disease Models, Animal , Humans , Mice , Osteosarcoma/pathology , Tumor Microenvironment
7.
Pharmacol Ther ; 188: 1-11, 2018 08.
Article in English | MEDLINE | ID: mdl-29476772

ABSTRACT

Extracellular vesicles (EVs) are heterogeneous multi-signal messengers that support cancer growth and dissemination by mediating the tumor-stroma crosstalk. Exosomes are a subtype of EVs that originate from the limiting membrane of late endosomes, and as such contain information linked to both the intrinsic cell "state" and the extracellular signals cells received from their environment. Resolving the signals affecting exosome biogenesis, cargo sorting and release will increase our understanding of tumorigenesis. In this review we highlight key cell biological processes that couple exosome biogenesis to cargo sorting in cancer cells. Moreover, we discuss how the bidirectional communication between tumor and non-malignant cells affect cancer growth and metastatic behavior.


Subject(s)
Extracellular Vesicles/physiology , Neoplasms/etiology , Animals , Cell Communication , Exosomes/physiology , Extracellular Matrix/physiology , Humans , Neoplasms/pathology , Stromal Cells/physiology , Tumor Microenvironment
8.
J Cell Biol ; 217(3): 1129-1142, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29339438

ABSTRACT

Exosomes are small endosome-derived extracellular vesicles implicated in cell-cell communication and are secreted by living cells when multivesicular bodies (MVBs) fuse with the plasma membrane (PM). Current techniques to study exosome physiology are based on isolation procedures after secretion, precluding direct and dynamic insight into the mechanics of exosome biogenesis and the regulation of their release. In this study, we propose real-time visualization of MVB-PM fusion to overcome these limitations. We designed tetraspanin-based pH-sensitive optical reporters that detect MVB-PM fusion using live total internal reflection fluorescence and dynamic correlative light-electron microscopy. Quantitative analysis demonstrates that MVB-PM fusion frequency is reduced by depleting the target membrane SNAREs SNAP23 and syntaxin-4 but also can be induced in single cells by stimulation of the histamine H1 receptor (H1HR). Interestingly, activation of H1R1 in HeLa cells increases Ser110 phosphorylation of SNAP23, promoting MVB-PM fusion and the release of CD63-enriched exosomes. Using this single-cell resolution approach, we highlight the modulatory dynamics of MVB exocytosis that will help to increase our understanding of exosome physiology and identify druggable targets in exosome-associated pathologies.


Subject(s)
Cell Membrane/physiology , Membrane Fusion/physiology , Multivesicular Bodies/physiology , Receptors, G-Protein-Coupled/metabolism , Cell Communication/drug effects , Cell Membrane/drug effects , Exocytosis/drug effects , HCT116 Cells , HeLa Cells , Histamine/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Membrane Fusion/drug effects , Multivesicular Bodies/drug effects , Phosphorylation/drug effects , Potassium Chloride/pharmacology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/metabolism , Receptors, G-Protein-Coupled/drug effects , Receptors, Histamine H1/drug effects , Single-Cell Analysis , Tetraspanins/genetics , Tetraspanins/metabolism
10.
Clin Cancer Res ; 23(14): 3721-3733, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28053020

ABSTRACT

Purpose: Human osteosarcoma is a genetically heterogeneous bone malignancy with poor prognosis despite the employment of aggressive chemotherapy regimens. Because druggable driver mutations have not been established, dissecting the interactions between osteosarcoma cells and supporting stroma may provide insights into novel therapeutic targets.Experimental Design: By using a bioluminescent orthotopic xenograft mouse model of osteosarcoma, we evaluated the effect of tumor extracellular vesicle (EV)-educated mesenchymal stem cells (TEMSC) on osteosarcoma progression. Characterization and functional studies were designed to assess the mechanisms underlying MSC education. Independent series of tissue specimens were analyzed to corroborate the preclinical findings, and the composition of patient serum EVs was analyzed after isolation with size-exclusion chromatography.Results: We show that EVs secreted by highly malignant osteosarcoma cells selectively incorporate a membrane-associated form of TGFß, which induces proinflammatory IL6 production by MSCs. TEMSCs promote tumor growth, accompanied with intratumor STAT3 activation and lung metastasis formation, which was not observed with control MSCs. Importantly, intravenous administration of the anti-IL6 receptor antibody tocilizumab abrogated the tumor-promoting effects of TEMSCs. RNA-seq analysis of human osteosarcoma tissues revealed a distinct TGFß-induced prometastatic gene signature. Tissue microarray immunostaining indicated active STAT3 signaling in human osteosarcoma, consistent with the observations in TEMSC-treated mice. Finally, we isolated pure populations of EVs from serum and demonstrated that circulating levels of EV-associated TGFß are increased in osteosarcoma patients.Conclusions: Collectively, our findings suggest that TEMSCs promote osteosarcoma progression and provide the basis for testing IL6- and TGFß-blocking agents as new therapeutic options for osteosarcoma patients. Clin Cancer Res; 23(14); 3721-33. ©2017 AACR.


Subject(s)
Interleukin-6/genetics , Lung Neoplasms/genetics , Osteosarcoma/genetics , STAT3 Transcription Factor/genetics , Transforming Growth Factor beta/genetics , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Extracellular Vesicles/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mesenchymal Stem Cells/metabolism , Mice , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Signal Transduction/genetics , Tissue Array Analysis
11.
JCI Insight ; 1(19): e89631, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27882350

ABSTRACT

BACKGROUND. Cell-free circulating nucleic acids, including 22-nt microRNAs (miRNAs), represent noninvasive biomarkers for treatment response monitoring of cancer patients. While the majority of plasma miRNA is bound to proteins, a smaller, less well-characterized pool is associated with extracellular vesicles (EVs). Here, we addressed whether EV-associated miRNAs reflect metabolic disease in classical Hodgkin lymphoma (cHL) patients. METHODS. With standardized size-exclusion chromatography (SEC), we isolated EV-associated extracellular RNA (exRNA) fractions and protein-bound miRNA from plasma of cHL patients and healthy subjects. We performed a comprehensive small RNA sequencing analysis and validation by TaqMan qRT-PCR for candidate discovery. Fluorodeoxyglucose-PET (FDG-PET) status before treatment, directly after treatment, and during long-term follow-up was compared directly with EV miRNA levels. RESULTS. The plasma EV miRNA repertoire was more extensive compared with protein-bound miRNA that was heavily dominated by a few abundant miRNA species and was less informative of disease status. Purified EV fractions of untreated cHL patients and tumor EVs had enriched levels of miR24-3p, miR127-3p, miR21-5p, miR155-5p, and let7a-5p compared with EV fractions from healthy subjects and disease controls. Serial monitoring of EV miRNA levels in patients before treatment, directly after treatment, and during long-term follow-up revealed robust, stable decreases in miRNA levels matching a complete metabolic response, as observed with FDG-PET. Importantly, EV miRNA levels rose again in relapse patients. CONCLUSION. We conclude that cHL-related miRNA levels in circulating EVs reflect the presence of vital tumor tissue and are suitable for therapy response and relapse monitoring in individual cHL patients. FUNDING. Cancer Center Amsterdam Foundation (CCA-2013), Dutch Cancer Society (KWF-5510), Technology Foundation STW (STW Perspectief CANCER-ID).


Subject(s)
Extracellular Vesicles , Hodgkin Disease/blood , MicroRNAs/blood , Biomarkers, Tumor/blood , Case-Control Studies , Hodgkin Disease/therapy , Humans , Neoplasm Recurrence, Local
12.
Proc Natl Acad Sci U S A ; 113(5): E587-96, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26768848

ABSTRACT

Complex interactions between DNA herpesviruses and host factors determine the establishment of a life-long asymptomatic latent infection. The lymphotropic Epstein-Barr virus (EBV) seems to avoid recognition by innate sensors despite massive transcription of immunostimulatory small RNAs (EBV-EBERs). Here we demonstrate that in latently infected B cells, EBER1 transcripts interact with the lupus antigen (La) ribonucleoprotein, avoiding cytoplasmic RNA sensors. However, in coculture experiments we observed that latent-infected cells trigger antiviral immunity in dendritic cells (DCs) through selective release and transfer of RNA via exosomes. In ex vivo tonsillar cultures, we observed that EBER1-loaded exosomes are preferentially captured and internalized by human plasmacytoid DCs (pDCs) that express the TIM1 phosphatidylserine receptor, a known viral- and exosomal target. Using an EBER-deficient EBV strain, enzymatic removal of 5'ppp, in vitro transcripts, and coculture experiments, we established that 5'pppEBER1 transfer via exosomes drives antiviral immunity in nonpermissive DCs. Lupus erythematosus patients suffer from elevated EBV load and activated antiviral immunity, in particular in skin lesions that are infiltrated with pDCs. We detected high levels of EBER1 RNA in such skin lesions, as well as EBV-microRNAs, but no intact EBV-DNA, linking non-cell-autonomous EBER1 presence with skin inflammation in predisposed individuals. Collectively, our studies indicate that virus-modified exosomes have a physiological role in the host-pathogen stand-off and may promote inflammatory disease.


Subject(s)
Dendritic Cells/virology , Epstein-Barr Virus Infections/genetics , Exosomes/metabolism , RNA, Viral/metabolism , Biological Transport , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/genetics , Humans , Proteome
SELECTION OF CITATIONS
SEARCH DETAIL
...