Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
1.
Cancers (Basel) ; 14(1)2022 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-35008424

RÉSUMÉ

With five-year survival rates as low as 3%, lung cancer is the most common cause of cancer-related mortality worldwide. The severity of the disease at presentation is accredited to the lack of early detection capacities, resulting in the reliance on low-throughput diagnostic measures, such as tissue biopsy and imaging. Interest in the development and use of liquid biopsies has risen, due to non-invasive sample collection, and the depth of information it can provide on a disease. Small extracellular vesicles (sEVs) as viable liquid biopsies are of particular interest due to their potential as cancer biomarkers. To validate the use of sEVs as cancer biomarkers, we characterised cancer sEVs using miRNA sequencing analysis. We found that miRNA-3182 was highly enriched in sEVs derived from the blood of patients with invasive breast carcinoma and NSCLC. The enrichment of sEV miR-3182 was confirmed in oncogenic, transformed lung cells in comparison to isogenic, untransformed lung cells. Most importantly, miR-3182 can successfully distinguish early-stage NSCLC patients from those with benign lung conditions. Therefore, miR-3182 provides potential to be used for the detection of NSCLC in blood samples, which could result in earlier therapy and thus improved outcomes and survival for patients.

2.
Cell ; 184(8): 2167-2182.e22, 2021 04 15.
Article de Anglais | MEDLINE | ID: mdl-33811809

RÉSUMÉ

Cardiac injury and dysfunction occur in COVID-19 patients and increase the risk of mortality. Causes are ill defined but could be through direct cardiac infection and/or inflammation-induced dysfunction. To identify mechanisms and cardio-protective drugs, we use a state-of-the-art pipeline combining human cardiac organoids with phosphoproteomics and single nuclei RNA sequencing. We identify an inflammatory "cytokine-storm", a cocktail of interferon gamma, interleukin 1ß, and poly(I:C), induced diastolic dysfunction. Bromodomain-containing protein 4 is activated along with a viral response that is consistent in both human cardiac organoids (hCOs) and hearts of SARS-CoV-2-infected K18-hACE2 mice. Bromodomain and extraterminal family inhibitors (BETi) recover dysfunction in hCOs and completely prevent cardiac dysfunction and death in a mouse cytokine-storm model. Additionally, BETi decreases transcription of genes in the viral response, decreases ACE2 expression, and reduces SARS-CoV-2 infection of cardiomyocytes. Together, BETi, including the Food and Drug Administration (FDA) breakthrough designated drug, apabetalone, are promising candidates to prevent COVID-19 mediated cardiac damage.


Sujet(s)
COVID-19/complications , Cardiotoniques/usage thérapeutique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Cardiopathies/traitement médicamenteux , Quinazolinones/usage thérapeutique , Facteurs de transcription/antagonistes et inhibiteurs , Angiotensin-converting enzyme 2/métabolisme , Animaux , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire , Cytokines/métabolisme , Femelle , Cardiopathies/étiologie , Cellules souches embryonnaires humaines , Humains , Inflammation/complications , Inflammation/traitement médicamenteux , Souris , Souris de lignée C57BL , Facteurs de transcription/métabolisme , Traitements médicamenteux de la COVID-19
3.
Immunol Cell Biol ; 99(1): 65-83, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32748462

RÉSUMÉ

Type 2 innate lymphoid cells (ILC2s) are important producers of type 2 cytokines whose role in hematological cancers remains unclear. ILC2s are a heterogeneous population encompassing distinct subsets with different tissue localization and cytokine responsiveness. In this study, we investigated the role of bone marrow (BM) ILC2s and interleukin (IL)-33-stimulated ILC2s in multiple myeloma, a plasma cell malignancy that develops in the BM. We found that myeloma growth was associated with phenotypic and functional alterations of BM ILC2s, characterized by an increased expression of maturation markers and reduced cytokine response to IL-2/IL-33. We identified a population of KLRG1hi ILC2s that preferentially accumulated in the liver and spleen of Il2rg-/- Rag2-/- mice reconstituted with BM ILC2s. A similar population of KLRG1hi ILC2s was observed in the blood, liver and spleen of IL-33-treated wild-type mice. The presence of KLRG1hi ILC2s in ILC2-reconstituted Il2rg-/- Rag2-/- mice or in IL-33-treated wild-type mice was associated with increased eosinophil numbers but had no effect on myeloma progression. Interestingly, while decreased myeloma growth was observed following treatment of Rag-deficient mice with the type 1 cytokines IL-12 and IL-18, this protection was reversed when mice received a combined treatment of IL-33 together with IL-12 and IL-18. In summary, our data indicate that IL-33 treatment induces a population of circulating inflammatory KLRG1hi ILC2s and inhibits type 1 immunity against multiple myeloma. These results argue against therapeutic administration of IL-33 to myeloma patients.


Sujet(s)
Immunité innée , Myélome multiple , Animaux , Cytokines , Humains , Interleukine-33 , Lectines de type C , Lymphocytes , Souris , Myélome multiple/traitement médicamenteux , Récepteurs immunologiques
4.
Immunity ; 53(4): 805-823.e15, 2020 10 13.
Article de Anglais | MEDLINE | ID: mdl-33053330

RÉSUMÉ

The activating receptor CD226 is expressed on lymphocytes, monocytes, and platelets and promotes anti-tumor immunity in pre-clinical models. Here, we examined the role of CD226 in the function of tumor-infiltrating lymphocytes (TILs) and resistance to immunotherapy. In murine tumors, a large proportion of CD8+ TILs had decreased surface expression of CD226 and exhibited features of dysfunction, whereas CD226hi TILs were highly functional. This correlation was seen also in TILs isolated from HNSCC patients. Mutation of CD226 at tyrosine 319 (Y319) led to increased CD226 surface expression, enhanced anti-tumor immunity and improved efficacy of immune checkpoint blockade (ICB). Mechanistically, tumor-derived CD155, the ligand for CD226, initiated phosphorylation of Y319 by Src kinases, thereby enabling ubiquitination of CD226 by CBL-B, internalization, and proteasomal degradation. In pre-treatment samples from melanoma patients, CD226+CD8+ T cells correlated with improved progression-free survival following ICB. Our findings argue for the development of therapies aimed at maintaining the expression of CD226.


Sujet(s)
Antigènes de différenciation des lymphocytes T/immunologie , Lymphocytes T CD8+/immunologie , Récepteurs viraux/immunologie , Animaux , Lignée cellulaire , Lignée cellulaire tumorale , Cellules HEK293 , Humains , Inhibiteurs de points de contrôle immunitaires/immunologie , Immunothérapie/méthodes , Cellules Jurkat , Lymphocytes TIL/immunologie , Mâle , Mélanome/immunologie , Souris , Souris de lignée C57BL
5.
JCI Insight ; 52019 06 13.
Article de Anglais | MEDLINE | ID: mdl-31194697

RÉSUMÉ

Immunotherapy holds promise for multiple myeloma (MM) patients but little is known about how MM-induced immunosuppression influences response to therapy. Here, we investigated the impact of disease progression on immunotherapy efficacy in the Vk*MYC mouse model. Treatment with agonistic anti-CD137 (4-1BB) mAbs efficiently protected mice when administered early but failed to contain MM growth when delayed more than three weeks after Vk*MYC tumor cell challenge. The quality of CD8+ T cell response to CD137 stimulation was not altered by the presence of MM, but CD8+ T cell numbers were profoundly reduced at the time of treatment. Our data suggest that an insufficient ratio of CD8+ T cells over MM cells (CD8/MM) accounts for the loss of anti-CD137 mAb efficacy. We established serum M-protein levels prior to therapy as a predictive factor of response. Moreover, we developed an in silico model to capture the dynamic interactions between CD8+ T cells and MM cells. Finally, we explored two methods to improve the CD8/MM ratio: anti-CD137 mAb immunotherapy combined with Treg-depletion or administered after chemotherapy treatment with cyclophosphamide or melphalan efficiently reduced MM burden and prolonged survival. Altogether, our data indicate that consolidation treatment with anti-CD137 mAbs might prevent MM relapse.


Sujet(s)
Ligand de 4-1BB/métabolisme , Anticorps monoclonaux/pharmacologie , Antinéoplasiques/pharmacologie , Immunothérapie/méthodes , Myélome multiple/traitement médicamenteux , Animaux , Anticorps monoclonaux/usage thérapeutique , Antigènes néoplasiques/immunologie , Antinéoplasiques/usage thérapeutique , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Modèles animaux de maladie humaine , Facteurs immunologiques/pharmacologie , Facteurs immunologiques/usage thérapeutique , Souris , Souris de lignée C57BL , Souris knockout , Myélome multiple/anatomopathologie , Lymphocytes T régulateurs
6.
Proteomics ; 19(8): e1800180, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30672117

RÉSUMÉ

A manner in which cells can communicate with each other is via secreted nanoparticles termed exosomes. These vesicles contain lipids, nucleic acids, and proteins, and are said to reflect the cell-of-origin. However, for the exosomal protein content, there is limited evidence in the literature to verify this statement. Here, proteomic assessment combined with pathway-enrichment analysis is used to demonstrate that the protein cargo of exosomes reflects the epithelial/mesenchymal phenotype of secreting breast cancer cells. Given that epithelial-mesenchymal plasticity is known to implicate various stages of cancer progression, the results suggest that breast cancer subtypes with distinct epithelial and mesenchymal phenotypes may be distinguished by directly assessing the protein content of exosomes. Additionally, the work is a substantial step toward verifying the statement that cell-derived exosomes reflect the phenotype of the cells-of-origin.


Sujet(s)
Tumeurs du sein/anatomopathologie , Animaux , Technique de Western , Tumeurs du sein/métabolisme , Tumeurs du sein/ultrastructure , Lignée cellulaire tumorale , Chromatographie en phase liquide , Transition épithélio-mésenchymateuse/physiologie , Exosomes/métabolisme , Exosomes/anatomopathologie , Exosomes/ultrastructure , Femelle , Humains , Spectrométrie de masse , Souris , Souris de lignée C57BL , Microscopie électronique à transmission
7.
Blood ; 132(16): 1689-1694, 2018 10 18.
Article de Anglais | MEDLINE | ID: mdl-29986909

RÉSUMÉ

Immune-based therapies hold promise for the treatment of multiple myeloma (MM), but so far, immune checkpoint blockade targeting programmed cell death protein 1 has not proven effective as single agent in this disease. T-cell immunoglobulin and ITIM domains (TIGIT) is another immune checkpoint receptor known to negatively regulate T-cell functions. In this study, we investigated the therapeutic potential of TIGIT blockade to unleash immune responses against MM. We observed that, in both mice and humans, MM progression was associated with high levels of TIGIT expression on CD8+ T cells. TIGIT+ CD8+ T cells from MM patients exhibited a dysfunctional phenotype characterized by decreased proliferation and inability to produce cytokines in response to anti-CD3/CD28/CD2 or myeloma antigen stimulation. Moreover, when challenged with Vk*MYC mouse MM cells, TIGIT-deficient mice showed decreased serum monoclonal immunoglobulin protein levels associated with reduced tumor burden and prolonged survival, indicating that TIGIT limits antimyeloma immune responses. Importantly, blocking TIGIT using monoclonal antibodies increased the effector function of MM patient CD8+ T cells and suppressed MM development. Altogether our data provide evidence for an immune-inhibitory role of TIGIT in MM and support the development of TIGIT-blocking strategies for the treatment of MM patients.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Lymphocytes T CD8+/immunologie , Myélome multiple/prévention et contrôle , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteurs immunologiques/antagonistes et inhibiteurs , Animaux , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Cellules cultivées , Humains , Activation des lymphocytes/immunologie , Souris , Souris de lignée C57BL , Souris knockout , Myélome multiple/étiologie , Myélome multiple/anatomopathologie , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/physiologie
8.
Front Immunol ; 9: 871, 2018.
Article de Anglais | MEDLINE | ID: mdl-29867925

RÉSUMÉ

Tumor-derived exosomes are being recognized as essential mediators of intercellular communication between cancer and immune cells. It is well established that bone marrow-derived macrophages (BMDMs) take up tumor-derived exosomes. However, the functional impact of these exosomes on macrophage phenotypes is controversial and not well studied. Here, we show that breast cancer-derived exosomes alter the phenotype of macrophages through the interleukin-6 (IL-6) receptor beta (glycoprotein 130, gp130)-STAT3 signaling pathway. Addition of breast cancer-derived exosomes to macrophages results in the activation of the IL-6 response pathway, including phosphorylation of the key downstream transcription factor STAT3. Exosomal gp130, which is highly enriched in cancer exosomes, triggers the secretion of IL-6 from BMDMs. Moreover, the exposure of BMDMs to cancer-derived exosomes triggers changes from a conventional toward a polarized phenotype often observed in tumor-associated macrophages. All of these effects can be inhibited through the addition of a gp130 inhibitor to cancer-derived exosomes or by blocking BMDMs exosome uptake. Collectively, this work demonstrates that breast cancer-derived exosomes are capable of inducing IL-6 secretion and a pro-survival phenotype in macrophages, partially via gp130/STAT3 signaling.


Sujet(s)
Exosomes/immunologie , Macrophages/immunologie , Tumeurs expérimentales de la mamelle/immunologie , Transduction du signal/immunologie , Microenvironnement tumoral/immunologie , Animaux , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Techniques de coculture , Récepteur gp130 de cytokines/antagonistes et inhibiteurs , Récepteur gp130 de cytokines/immunologie , Récepteur gp130 de cytokines/métabolisme , Exosomes/effets des médicaments et des substances chimiques , Exosomes/métabolisme , Femelle , Hydrazines/pharmacologie , Interleukine-6/immunologie , Interleukine-6/métabolisme , Activation des macrophages/effets des médicaments et des substances chimiques , Activation des macrophages/immunologie , Macrophages/cytologie , Macrophages/métabolisme , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris de lignée C57BL , Culture de cellules primaires , Quinoxalines/pharmacologie , Facteur de transcription STAT-3/immunologie , Facteur de transcription STAT-3/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
9.
PLoS One ; 13(4): e0196040, 2018.
Article de Anglais | MEDLINE | ID: mdl-29677215

RÉSUMÉ

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid progenitor cells that are expanded in cancer and act as potent suppressors of the anti-tumor immune response. MDSCs consist of two major subsets, namely monocytic (M-) MDSCs and granulocytic (G-) MDSCs that differ with respect to their phenotype, morphology and mechanisms of suppression. Here, we cultured bone marrow cells with IL-6 and GM-CSF in vitro to generate a population of bone marrow MDSCs (BM-MDSCs) similar to G-MDSCs from tumor-bearing mice in regards to phenotype, morphology and suppressive-function. Through fluorescent labeling of these BM-MDSCs and optical imaging, we could visualize the recruitment and localization of BM-MDSCs in breast tumor-bearing mice in vivo. Furthermore, we were able to demonstrate that BM-MDSCs home to primary and metastatic breast tumors, but have no significant effect on tumor growth or progression. Ex vivo flow cytometry characterization of BM-MDSCs after adoptive transfer demonstrated both organ-and tumor-specific effects on their phenotype and differentiation, demonstrating the importance of the local microenvironment on MDSC fate and function. In this study, we have developed a method to generate, visualize and detect BM-MDSCs in vivo and ex vivo through optical imaging and flow cytometry, in order to understand the organ-specific changes rendered to MDSCs in breast cancer.


Sujet(s)
Transfert adoptif/méthodes , Facteur de stimulation des colonies de granulocytes et de macrophages/pharmacologie , Interleukine-6/pharmacologie , Tumeurs expérimentales de la mamelle/imagerie diagnostique , Cellules myéloïdes suppressives/cytologie , Animaux , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/immunologie , Techniques de culture cellulaire , Lignée cellulaire tumorale , Cellules cultivées , Femelle , Activation des lymphocytes , Tumeurs expérimentales de la mamelle/immunologie , Souris , Cellules myéloïdes suppressives/immunologie , Métastase tumorale , Imagerie optique , Transplantation de cellules souches , Microenvironnement tumoral
10.
Cancer Res ; 76(23): 6816-6827, 2016 12 01.
Article de Anglais | MEDLINE | ID: mdl-27760789

RÉSUMÉ

Small membranous secretions from tumor cells, termed exosomes, contribute significantly to intercellular communication and subsequent reprogramming of the tumor microenvironment. Here, we use optical imaging to determine that exogenously administered fluorescently labeled exosomes derived from highly metastatic murine breast cancer cells distributed predominantly to the lung of syngeneic mice, a frequent site of breast cancer metastasis. At the sites of accumulation, exosomes were taken up by CD45+ bone marrow-derived cells. Subsequent long-term conditioning of naïve mice with exosomes from highly metastatic breast cancer cells revealed the accumulation of myeloid-derived suppressor cells in the lung and liver. This favorable immune suppressive microenvironment was capable of promoting metastatic colonization in the lung and liver, an effect not observed from exosomes derived from nonmetastatic cells and liposome control vesicles. Furthermore, we determined that breast cancer exosomes directly suppressed T-cell proliferation and inhibited NK cell cytotoxicity, and hence likely suppressed the anticancer immune response in premetastatic organs. Together, our findings provide novel insight into the tissue-specific outcomes of breast cancer-derived exosome accumulation and their contribution to immune suppression and promotion of metastases. Cancer Res; 76(23); 6816-27. ©2016 AACR.


Sujet(s)
Tumeurs du sein/immunologie , Exosomes/métabolisme , Immunosuppression thérapeutique/méthodes , Animaux , Tumeurs du sein/anatomopathologie , Prolifération cellulaire , Femelle , Humains , Souris , Souris de lignée BALB C , Souris de lignée C57BL
11.
EMBO Mol Med ; 5(12): 1835-51, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-24142880

RÉSUMÉ

Muscle-invasive forms of urothelial carcinomas are responsible for most mortality in bladder cancer. Finding new treatments for invasive bladder tumours requires adequate animal models to decipher the mechanisms of progression, in particular the way tumours interact with their microenvironment. Herein, using the murine bladder tumour cell line MB49 and its more aggressive variant MB49-I, we demonstrate that the adaptive immune system efficiently limits progression of MB49, whereas MB49-I has lost tumour antigens and is insensitive to adaptive immune responses. Furthermore, we unravel a parallel mechanism developed by MB49-I to subvert its environment: de novo secretion of the proteoglycan decorin. We show that decorin overexpression in the MB49/MB49-I model is required for efficient progression, by promoting angiogenesis and tumour cell invasiveness. Finally, we show that these results are relevant to muscle-invasive human bladder carcinomas, which overexpress decorin together with angiogenesis- and adhesion/migration-related genes, and that decorin overexpression in the human bladder carcinoma cell line TCCSUP is required for efficient invasiveness in vitro. We thus propose decorin as a new therapeutic target for these aggressive tumours.


Sujet(s)
Décorine/métabolisme , Immunité acquise , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire , Cytokines/métabolisme , Protéines de liaison à l'ADN/déficit , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Décorine/antagonistes et inhibiteurs , Décorine/génétique , Femelle , Humains , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Interférence par ARN , Petit ARN interférent/métabolisme , Lymphocytes T/cytologie , Lymphocytes T/immunologie , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/anatomopathologie
12.
Neoplasia ; 15(1): 85-94, 2013 Jan.
Article de Anglais | MEDLINE | ID: mdl-23359264

RÉSUMÉ

Chemotherapy enhances the antitumor adaptive immune T cell response, but the immunosuppressive tumor environment often dominates, resulting in cancer relapse. Antigen-presenting cells such as tumor-associated macrophages (TAMs) and tumor dendritic cells (TuDCs) are the main protagonists of tumor-infiltrating lymphocyte (TIL) immunosuppression. TAMs have been widely investigated and are associated with poor prognosis, but the immunosuppressive activity of TuDCs is less well understood. We performed two-photon imaging of the tumor tissue to examine the spatiotemporal interactions between TILs and TuDCs after chemotherapy. In a strongly immunosuppressive murine tumor model, cyclophosphamide-mediated chemotherapy transiently enhanced the antitumor activity of adoptively transferred ovalbumin-specific CD8(+) T cell receptor transgenic T cells (OTI) but barely affected TuDC compartment within the tumor. Time lapse imaging of living tumor tissue showed that TuDCs are organized as a mesh with dynamic interconnections. Once infiltrated into the tumor parenchyma, OTI T cells make antigen-specific and long-lasting contacts with TuDCs. Extensive analysis of TIL infiltration on histologic section revealed that after chemotherapy the majority of OTI T cells interact with TuDCs and that infiltration is restricted to TuDC-rich areas. We propose that the TuDC network exerts antigen-dependent unproductive retention that trap T cells and limit their antitumor effectiveness.


Sujet(s)
Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Lymphocytes TIL/immunologie , Lymphocytes T cytotoxiques/immunologie , Animaux , Antigènes/immunologie , Femelle , Tolérance immunitaire/immunologie , Souris , Souris de lignée C57BL , Souris transgéniques/immunologie , Récepteurs aux antigènes des cellules T/immunologie
13.
Cancer Res ; 72(19): 4920-30, 2012 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-22865453

RÉSUMÉ

During progression from single cancer cells to a tumor mass and metastases, tumor cells send signals that can subvert their tissue microenvironment. These signals involve soluble molecules and various extracellular vesicles, including a particular type termed exosomes. The specific roles of exosomes secreted in the tumor microenvironment, however, is unclear. The small GTPases RAB27A and RAB27B regulate exocytosis of multivesicular endosomes, which lead to exosome secretion, in human HeLa cells. Here, we used mouse models to show that Rab27a blockade in mammary carcinoma cells decreased secretion of exosomes characterized by endocytic markers, but also of matrix metalloproteinase 9, which is not associated with exosomes. Rab27a blockade resulted in decreased primary tumor growth and lung dissemination of a metastatic carcinoma (4T1), but not of a nonmetastatic carcinoma (TS/A). Local growth of 4T1 tumors required mobilization of a population of neutrophil immune cells induced by Rab27a-dependent secretion of exosomes together with a specific combination of cytokines and/or metalloproteinases. Our findings offer in vivo validation of the concept that exosome secretion can exert key pathophysiologic roles during tumor formation and progression, but they also highlight the idiosyncratic character of the tumor context.


Sujet(s)
Exosomes/métabolisme , Tumeurs expérimentales de la mamelle/métabolisme , Microenvironnement tumoral , Protéines G rab/métabolisme , Animaux , Technique de Western , Lignée cellulaire tumorale , Chimiokines/métabolisme , Cytokines/métabolisme , Évolution de la maladie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs expérimentales de la mamelle/génétique , Tumeurs expérimentales de la mamelle/anatomopathologie , Secreted matrix metalloproteinases/métabolisme , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/anatomopathologie , Interférence par ARN , RT-PCR , Charge tumorale/génétique , Protéines G rab/génétique , Protéines rab27 liant le GTP
14.
PLoS One ; 7(3): e33244, 2012.
Article de Anglais | MEDLINE | ID: mdl-22438901

RÉSUMÉ

PURPOSE: Milk fat globule-epidermal growth factor-factor VIII (MFGE8) is necessary for diurnal outer segment phagocytosis and promotes VEGF-dependent neovascularization. The prevalence of two single nucleotide polymorphisms (SNP) in MFGE8 was studied in two exsudative or "wet" Age-related Macular Degeneration (AMD) groups and two corresponding control groups. We studied the effect of MFGE8 deficiency on retinal homeostasis with age and on choroidal neovascularization (CNV) in mice. METHODS: The distribution of the SNP (rs4945 and rs1878326) of MFGE8 was analyzed in two groups of patients with "wet" AMD and their age-matched controls from Germany and France. MFGE8-expressing cells were identified in Mfge8(+/-) mice expressing ß-galactosidase. Aged Mfge8(+/-) and Mfge8(-/-) mice were studied by funduscopy, histology, electron microscopy, scanning electron microscopy of vascular corrosion casts of the choroid, and after laser-induced CNV. RESULTS: rs1878326 was associated with AMD in the French and German group. The Mfge8 promoter is highly active in photoreceptors but not in retinal pigment epithelium cells. Mfge8(-/-) mice did not differ from controls in terms of fundus appearance, photoreceptor cell layers, choroidal architecture or laser-induced CNV. In contrast, the Bruch's membrane (BM) was slightly but significantly thicker in Mfge8(-/-) mice as compared to controls. CONCLUSIONS: Despite a reproducible minor increase of rs1878326 in AMD patients and a very modest increase in BM in Mfge8(-/-) mice, our data suggests that MFGE8 dysfunction does not play a critical role in the pathogenesis of AMD.


Sujet(s)
Antigènes de surface/génétique , Antigènes de surface/physiologie , Néovascularisation choroïdienne/étiologie , Protéines de lait/génétique , Dégénérescence maculaire humide/étiologie , Sujet âgé , Animaux , Séquence nucléotidique , Études cas-témoins , Choroïde/physiologie , Néovascularisation choroïdienne/génétique , Néovascularisation choroïdienne/physiopathologie , Amorces ADN/génétique , Femelle , Expression des gènes , Homéostasie , Humains , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Polymorphisme de nucléotide simple , Rétine/physiologie , Dégénérescence maculaire humide/génétique , Dégénérescence maculaire humide/physiopathologie
15.
Article de Anglais | MEDLINE | ID: mdl-24009879

RÉSUMÉ

Exosomes are extracellular vesicles of 50 to 100 nm in diameter, released by many cell types. Exosomes are formed inside the cell in intracellular endosomal compartments and are secreted upon fusion of these compartments with the plasma membrane. Cells also secrete other types of membrane vesicles, for instance, by outward budding from the plasma membrane, and although some of them clearly differ from exosomes by their structural features (larger size), others are possibly more difficult to separate. Here, using Rab27a inhibition to modulate exosome secretion, we show the existence of at least 2 distinct populations of vesicles after purification by classical ultracentrifugation from mouse tumor cell conditioned medium. Rab27a inhibition lead to decreased vesicular secretion of some conventional markers of exosomes (CD63, Tsg101, Alix and Hsc70) but did not affect secretion of others (CD9 and Mfge8). By electron microscopy, CD9 was observed on vesicles of various sizes, ranging from 30 nm to more than 150 nm in diameter. Flotation onto sucrose gradients showed different proportions of CD63, CD9 and Mfge8 not only in fractions of densities classically described for exosomes (around 1.15 g/ml) but also in fractions of densities over 1.20 g/ml, indicating the presence of heterogenous vesicle populations. CD9 and Mfge8 were also found in large vesicles pelleted at low speed and can thus not be considered as specific components of endosome-derived vesicles. We propose that the most commonly used protocols for exosome preparations co-purify vesicles from endosomal and other origins, possibly the plasma membrane. Future work will be required to improve techniques for accurate purification and characterization of the different populations of extracellular vesicles.

16.
Nat Cell Biol ; 12(1): 19-30; sup pp 1-13, 2010 Jan.
Article de Anglais | MEDLINE | ID: mdl-19966785

RÉSUMÉ

Exosomes are secreted membrane vesicles that share structural and biochemical characteristics with intraluminal vesicles of multivesicular endosomes (MVEs). Exosomes could be involved in intercellular communication and in the pathogenesis of infectious and degenerative diseases. The molecular mechanisms of exosome biogenesis and secretion are, however, poorly understood. Using an RNA interference (RNAi) screen, we identified five Rab GTPases that promote exosome secretion in HeLa cells. Among these, Rab27a and Rab27b were found to function in MVE docking at the plasma membrane. The size of MVEs was strongly increased by Rab27a silencing, whereas MVEs were redistributed towards the perinuclear region upon Rab27b silencing. Thus, the two Rab27 isoforms have different roles in the exosomal pathway. In addition, silencing two known Rab27 effectors, Slp4 (also known as SYTL4, synaptotagmin-like 4) and Slac2b (also known as EXPH5, exophilin 5), inhibited exosome secretion and phenocopied silencing of Rab27a and Rab27b, respectively. Our results therefore strengthen the link between MVEs and exosomes, and introduce ways of manipulating exosome secretion in vivo.


Sujet(s)
Communication cellulaire , Endosomes/physiologie , Exosomes/métabolisme , Protéines G rab/métabolisme , Membrane cellulaire/métabolisme , Cytométrie en flux , Technique d'immunofluorescence , Extinction de l'expression des gènes , Cellules HeLa , Humains , Immunotransfert , ARN messager/génétique , ARN messager/métabolisme , Petit ARN interférent/pharmacologie , RT-PCR , Fractions subcellulaires , Protéines du transport vésiculaire/antagonistes et inhibiteurs , Protéines du transport vésiculaire/génétique , Protéines du transport vésiculaire/métabolisme , Protéines G rab/antagonistes et inhibiteurs , Protéines G rab/génétique , Protéines rab27 liant le GTP
17.
J Neuroimmunol ; 210(1-2): 120-3, 2009 May 29.
Article de Anglais | MEDLINE | ID: mdl-19278738

RÉSUMÉ

We investigated an association of the HLA-A locus in 78 French Caucasian patients with autoimmune myasthenia gravis (MG) and thymic epithelial tumours. The largest effect was a protection associated with HLA-A02 in MG patients with a B2 type thymoma (OR=0.323, 95% CI: 0.113-0.756, P=0.00041). The frequency of HLA-A25 was also increased in the whole group of patients (OR=3.62, 95% CI: 1.62-7.08, P=0.0041). Our findings emphasise the interest of the histological classification in the genetic study of thymomas.


Sujet(s)
Prédisposition génétique à une maladie/génétique , Antigènes HLA-A/génétique , Myasthénie/génétique , Myasthénie/immunologie , Thymome/génétique , Thymome/immunologie , Adulte , Autoanticorps/analyse , Autoanticorps/sang , Analyse de mutations d'ADN , Femelle , France , Fréquence d'allèle , Marqueurs génétiques/génétique , Dépistage génétique , Génétique des populations , Génotype , Hétérozygote , Test d'histocompatibilité , Humains , Mâle , Myasthénie/ethnologie , Thymome/anatomopathologie , Thymus (glande)/immunologie , Thymus (glande)/anatomopathologie , Thymus (glande)/physiopathologie ,
18.
Cancer Res ; 68(4): 1228-35, 2008 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-18281500

RÉSUMÉ

Expression of non-self antigens by tumors can induce activation of T cells in vivo, although this activation can lead to either immunity or tolerance. CD8+ T-cell activation can be direct (if the tumor expresses MHC class I molecules) or indirect (after the capture and cross-presentation of tumor antigens by dendritic cells). The modes of tumor antigen capture by dendritic cells in vivo remain unclear. Here we examine the immunogenicity of the same model antigen secreted by live tumors either in association with membrane vesicles (exosomes) or as a soluble protein. We have artificially addressed the antigen to secreted vesicles by coupling it to the factor VIII-like C1C2 domain of milk fat globule epidermal growth factor-factor VIII (MFG-E8)/lactadherin. We show that murine fibrosarcoma tumor cells that secrete vesicle-bound antigen grow slower than tumors that secrete soluble antigen in immunocompetent, but not in immunodeficient, host mice. This growth difference is due to the induction of a more potent antigen-specific antitumor immune response in vivo by the vesicle-bound than by the soluble antigen. Finally, in vivo secretion of the vesicle-bound antigen either by tumors or by vaccination with naked DNA protects against soluble antigen-secreting tumors. We conclude that the mode of secretion can determine the immunogenicity of tumor antigens and that manipulation of the mode of antigen secretion may be used to optimize antitumor vaccination protocols.


Sujet(s)
Antigènes néoplasiques/immunologie , Fibrosarcome/immunologie , Vésicules de sécrétion/immunologie , Animaux , Antigènes de surface/immunologie , Lymphocytes T CD8+/immunologie , Vaccins anticancéreux/génétique , Vaccins anticancéreux/immunologie , Embryon de poulet , Fibrosarcome/métabolisme , Souris , Souris de lignée C57BL , Protéines de lait/immunologie , Ovalbumine/génétique , Ovalbumine/immunologie , Ovalbumine/métabolisme , Vaccins à ADN/génétique , Vaccins à ADN/immunologie
19.
Ann Neurol ; 59(2): 404-7, 2006 Feb.
Article de Anglais | MEDLINE | ID: mdl-16437561

RÉSUMÉ

OBJECTIVE: Our objective was to investigate a role of the intracellular tyrosine phosphatase PTPN22*R620W variant in autoimmune myasthenia gravis (MG), considering disease heterogeneity. METHODS: We used a case-control design, comparing 470 patients and 296 controls, all French whites. Patients were categorized depending on the presence of a thymoma and serum anti-titin antibodies. RESULTS: The 620W risk allele was increased in 293 nonthymoma patients without anti-titin antibodies (odds ratio, 1.97; 95% confidence interval, 1.32-2.97, p = 0.00059) but not in nonthymoma patients with anti-titin antibodies or in thymoma patients. INTERPRETATION: Our genetic findings strengthen the concept that these groups of patients correspond to etiologically distinct disease entities.


Sujet(s)
Arginine/génétique , Prédisposition génétique à une maladie , Myasthénie/génétique , Polymorphisme génétique , Protein Tyrosine Phosphatases/génétique , Tryptophane/génétique , Adulte , Sujet âgé , Allèles , Intervalles de confiance , Connectine , Analyse de mutations d'ADN/méthodes , Femelle , Fréquence d'allèle , Humains , Mâle , Adulte d'âge moyen , Protéines du muscle/métabolisme , Myasthénie/classification , Odds ratio , Protein kinases/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 22
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...