Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Microsc Res Tech ; 87(8): 1733-1741, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38501548

ABSTRACT

The peripheral nerve injury (PNI) affects the morphology of the whole locomotor apparatus, which can reach the myotendinous junction (MTJ) interface. In the injury condition, the skeletal muscle satellite cells (SC) are triggered, activated, and proliferated to repair their structure, and in the MTJ, the telocytes (TC) are associated to support the interface with the need for remodeling; in that way, these cells can be associated with SC. The study aimed to describe the SC and TC relationship after PNI at the MTJ. Sixteen adult Wistar rats were divided into Control Group (C, n = 8) and PNI Group (PNI, n = 8), PNI was performed by the constriction of the sciatic nerve. The samples were processed for transmission electron microscopy and immunostaining analysis. In the C group was evidenced the arrangement of sarcoplasmic evaginations and invaginations, the support collagen layer with a TC inside it, and an SC through vesicles internally and externally to then. In the PNI group were observed the disarrangement of invaginations and evaginations and sarcomeres degradation at MTJ, as the disposition of telopodes adjacent and in contact to the SC with extracellular vesicles and exosomes in a characterized paracrine activity. These findings can determine a link between the TCs and the SCs at the MTJ remodeling. RESEARCH HIGHLIGHTS: Peripheral nerve injury promotes the myotendinous junction (MTJ) remodeling. The telocytes (TC) and the satellite cells (SC) are present at the myotendinous interface. TC mediated the SC activity at MTJ.


Subject(s)
Extracellular Vesicles , Microscopy, Electron, Transmission , Rats, Wistar , Satellite Cells, Skeletal Muscle , Telocytes , Animals , Telocytes/physiology , Telocytes/ultrastructure , Satellite Cells, Skeletal Muscle/physiology , Satellite Cells, Skeletal Muscle/cytology , Rats , Extracellular Vesicles/ultrastructure , Extracellular Vesicles/metabolism , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/metabolism , Male , Sciatic Nerve/ultrastructure , Tendons/physiology , Muscle, Skeletal/ultrastructure , Myotendinous Junction
2.
Arq. bras. med. vet. zootec. (Online) ; 75(1): 147-152, 2023. ilus, tab, graf
Article in English | VETINDEX | ID: biblio-1416627

ABSTRACT

As vesículas extracelulares (VEs) são nanoparticulas circundadas por uma bicamada lipídica, que transportam lipídios, proteínas, ácidos nucleicos e metabólitos, e podem ser secretadas por diferentes tipos celulares. Os adipócitos são capazes de sintetizar e secretar VEs pequenas, que, uma vez na circulação, podem alcançar diferentes sistemas orgânicos, permitindo a comunicação intercelular. O objetivo deste estudo foi avaliar o diâmetro e a concentração de VEs pequenas no soro de cães com sobrepeso e obesidade, antes e após a perda de peso. Para isso, foram recrutados 19 cães, sendo nove fêmeas e 10 machos castrados, em bom estado geral e com escore de condição corporal (ECC) ≥ 7. A avaliação inicial dos cães incluiu exame físico e testes laboratoriais. Após avaliação inicial, os cães foram alimentados com uma dieta hipocalórica comercial por quatro meses, e, após esse período, os testes laboratoriais foram reavaliados. As VEs pequenas foram isoladas a partir do sangue total, por meio de centrifugações e ultracentrifugações seriadas, e o equipamento NanoSight foi utilizado para a determinação do diâmetro e da concentração das VEs pequenas presentes no soro antes e após a dieta. Houve uma redução de 10% do peso corporal e de 16,5% do ECC dos cães, além da diminuição de 9% do diâmetro e de 31% da concentração das VEs pequenas, após os quatro meses de alimentação com a dieta hipocalórica. Não foram observadas diferenças em relação às análises bioquímicas e entre os resultados de fêmeas e machos, antes e após a perda de peso. Em conclusão, a redução do peso corporal e do acúmulo de gordura foi capaz de modificar as características (diâmetro e concentração) das VEs pequenas, que foram isoladas a partir do soro de cães com sobrepeso e obesidade.


Subject(s)
Animals , Dogs , Weight Loss/physiology , Overweight/veterinary , Extracellular Vesicles/ultrastructure , Obesity/veterinary , Caloric Restriction/veterinary , Liquid Biopsy/veterinary
3.
Cell Mol Life Sci ; 79(1): 11, 2021 Dec 24.
Article in English | MEDLINE | ID: mdl-34951683

ABSTRACT

Trichomonas vaginalis is a common sexually transmitted extracellular parasite that adheres to epithelial cells in the human urogenital tract. Extracellular vesicles (EVs) have been described as important players in the pathogenesis of this parasite as they deliver proteins and RNA into host cells and modulate parasite adherence. EVs are heterogeneous membrane vesicles released from virtually all cell types that collectively represent a new dimension of intercellular communication. The Endosomal Sorting Complex Required for Transport (ESCRT) machinery contributes to several key mechanisms in which it reshapes membranes. Based on this, some components of the ESCRT have been implicated in EVs biogenesis in other cells. Here, we demonstrated that VPS32, a member of ESCRTIII complex, contribute to the biogenesis and cargo sorting of extracellular vesicles in the parasite T. vaginalis. Moreover, we observe that parasites overexpressing VPS32 have a striking increase in adherence to host cells compared to control parasites; demonstrating a key role for this protein in mediating host: parasite interactions. These results provide valuable information on the molecular mechanisms involved in extracellular vesicles biogenesis, cargo-sorting, and parasite pathogenesis.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Extracellular Vesicles/metabolism , Host-Parasite Interactions , Parasites/cytology , Trichomonas vaginalis/cytology , Animals , Cell Adhesion , Cell Line , Extracellular Vesicles/ultrastructure , Humans , Male , Parasites/metabolism , Prostate/parasitology , Tandem Mass Spectrometry , Trichomonas vaginalis/metabolism
4.
Microbiol Spectr ; 9(1): e0012521, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34346749

ABSTRACT

Extracellular vesicles (EVs) produced by members of the Cryptococcus genus are associated with fundamental processes of fungal physiology and virulence. However, several questions about the properties of cryptococcal EVs remain unanswered, mostly because of technical limitations. We recently described a fast and efficient protocol of high-yield EV isolation from solid medium. In this study, we aimed at using the solid medium protocol to address some of the open questions about EVs, including the kinetics of EV production, the diversity of EVs produced by multiple isolates under different culture conditions, the separation of vesicles in a density gradient followed by the recovery of functional EVs, the direct detection of EVs in culture supernatants, and the production of vesicles in solid cultures of Titan cells. Our results indicate that the production of EVs is directly impacted by the culture medium and time of growth, resulting in variable detection of EVs per cell and a peak of EV detection at 24 h of growth. Nanoparticle tracking analysis (NTA) of EV samples revealed that multiple isolates produce vesicles with variable properties, including particles of diverging dimensions. EVs were produced in the solid medium in amounts that were separated on a centrifugation density gradient, resulting in the recovery of functional EVs containing the major cryptococcal capsular antigen. We also optimized the solid medium protocol for induction of the formation of Titan cells, and analyzed the production of EVs by NTA and transmission electron microscopy. This analysis confirmed that EVs were isolated from solid cultures of cryptococcal enlarged cells. With these approaches, we expect to implement simple methods that will facilitate the analysis of EVs produced by fungal cells. IMPORTANCE Fungal extracellular vesicles (EVs) are considered to be important players in the biology of fungal pathogens. However, the limitations in the methodological approaches to studying fungal EVs impair the expansion of knowledge in this field. In the present study, we used the Cryptococcus genus as a model for the study of EVs. We explored the simplification of protocols for EV analysis, which helped us to address some important, but still unanswered, questions about fungal EVs.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus/chemistry , Extracellular Vesicles/chemistry , Cryptococcus/classification , Cryptococcus/genetics , Cryptococcus/isolation & purification , Culture Media/chemistry , Culture Media/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Humans , Kinetics , Microscopy, Electron, Transmission
5.
Toxicol Lett ; 347: 12-22, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33945863

ABSTRACT

p-Cresyl sulfate (PCS), indoxyl sulfate (IS), and inorganic phosphate (Pi) are uremic toxins found in chronic kidney disease (CKD) that are closely related to endothelial extracellular vesicles (EVs) formation. The present study aimed to understand the role of EVs and their role in cell adhesion and migration, inflammation, and oxidative stress. Human endothelial cells were treated with PCS, IS, and Pi in pre-established uremic and kinetic recommendations. EVs were characterized using scanning electron microscopy, flow cytometry, and NanoSight assays. The concentrations of EVs were established using Alamar Blue and MTT assays. Cell adhesion to extracellular matrix proteins was analyzed using an adhesion assay. Inflammation and oxidative stress were assessed by vascular cell adhesion molecule-1 expression/monocyte migration and reactive oxygen species production, respectively. The capacity of EVs to stimulate endothelial cell migration was evaluated using a wound-healing assay. Our data showed that endothelial cells stimulated with uremic toxins can induce the formation of EVs of different sizes, quantities, and concentrations, depending on the uremic toxin used. Cell adhesion was significantly (P < 0.01) stimulated in cells exposed to PCS-induced extracellular vesicles (PCSEVs) and inorganic phosphate-induced extracellular vesicles (PiEVs). Cell migration was significantly (P < 0.05) stimulated by PCSEVs. VCAM-1 expression was evident in cells treated with PCSEVs and IS-induced extracellular vesicles (ISEVs). EVs are not able to stimulate monocyte migration or oxidative stress. In conclusion, EVs may be a biomarker of endothelial injury and the inflammatory process, playing an important role in cell-to-cell communication and pathophysiological processes, although more studies are needed to better understand the mechanisms of EVs in uremia.


Subject(s)
Cell Adhesion/drug effects , Cell Movement/drug effects , Cresols/toxicity , Endothelial Cells/drug effects , Extracellular Vesicles/drug effects , Indican/toxicity , Inflammation Mediators/metabolism , Oxidative Stress/drug effects , Phosphates/toxicity , Sulfuric Acid Esters/toxicity , Uremia/pathology , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Humans , Signal Transduction , Uremia/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
6.
Front Immunol ; 12: 654574, 2021.
Article in English | MEDLINE | ID: mdl-33796117

ABSTRACT

The small molecule (molecular mass <900 Daltons) composition of extracellular vesicles (EVs) produced by the pathogenic fungus Cryptococcus gattii is unknown, which limits the understanding of the functions of cryptococcal EVs. In this study, we analyzed the composition of small molecules in samples obtained from solid cultures of C. gattii by a combination of chromatographic and spectrometric approaches, and untargeted metabolomics. This analysis revealed previously unknown components of EVs, including small peptides with known biological functions in other models. The peptides found in C. gattii EVs had their chemical structure validated by chemical approaches and comparison with authentic standards, and their functions tested in a Galleria mellonella model of cryptococcal infection. One of the vesicular peptides (isoleucine-proline-isoleucine, Ile-Pro-Ile) improved the survival of G. mellonella lethally infected with C. gattii or C. neoformans. These results indicate that small molecules exported in EVs are biologically active in Cryptococcus. Our study is the first to characterize a fungal EV molecule inducing protection, pointing to an immunological potential of extracellular peptides produced by C. gattii.


Subject(s)
Cryptococcosis/metabolism , Cryptococcus gattii/physiology , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Host-Pathogen Interactions , Invertebrates , Animals , Computational Biology/methods , Cryptococcosis/microbiology , Extracellular Vesicles/ultrastructure , Metabolomics/methods , Molecular Structure , Peptides
7.
Sci Rep ; 10(1): 15160, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32939048

ABSTRACT

Genetically modified tumor cells harboring immunomodulators may be used as therapeutic vaccines to stimulate antitumor immunity. The therapeutic benefit of these tumor vaccines is extensively investigated and mechanisms by which they boost antitumor response may be further explored. Tumor cells are large secretors of extracellular vesicles (EVs). These EVs are able to vehiculate RNA and proteins to target cells, and engineered EVs also vehiculate recombinant proteins. In this study, we explore immunomodulatory properties of EVs derived from antitumor vaccines expressing the TNFSF ligands 4-1BBL and OX40L, modulating immune response mediated by immune cells and eliminating tumors. Our results suggest that the EVs secreted by genetically modified tumor cells harboring TNFSF ligands can induce T cell proliferation, inhibit the transcription factor FoxP3, associated with the maintenance of Treg phenotype, and enhance antitumor activity mediated by immune cells. The immunomodulatory extracellular vesicles have potential to be further engineered for developing new approaches for cancer therapy.


Subject(s)
4-1BB Ligand/immunology , Cancer Vaccines/therapeutic use , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , OX40 Ligand/immunology , 4-1BB Ligand/genetics , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Extracellular Vesicles/genetics , Extracellular Vesicles/immunology , Extracellular Vesicles/ultrastructure , Forkhead Transcription Factors/antagonists & inhibitors , Immunologic Factors/genetics , Immunologic Factors/immunology , Immunologic Factors/therapeutic use , In Vitro Techniques , Lymphocyte Activation , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , OX40 Ligand/genetics
8.
Cell Commun Signal ; 18(1): 158, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32988382

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs) are lipid-bound particles that are naturally released from cells and mediate cell-cell communication. Integrin adhesion receptors are enriched in small EVs (SEVs) and SEV-carried integrins have been shown to promote cancer cell migration and to mediate organ-specific metastasis; however, how integrins mediate these effects is not entirely clear and could represent a combination of EV binding to extracellular matrix and cells. METHODS: To probe integrin role in EVs binding and uptake, we employed a disintegrin inhibitor (DisBa-01) of integrin binding with specificity for αvß3 integrin. EVs were purified from MDA-MB-231 cells conditioned media by serial centrifugation method. Isolated EVs were characterized by different techniques and further employed in adhesion, uptake and co-culture experiments. RESULTS: We find that SEVs secreted from MDA-MB-231 breast cancer cells carry αvß3 integrin and bind directly to fibronectin-coated plates, which is inhibited by DisBa-01. SEV coating on tissue culture plates also induces adhesion of MDA-MB-231 cells, which is inhibited by DisBa-01 treatment. Analysis of EV uptake and interchange between cells reveals that the amount of CD63-positive EVs delivered from malignant MDA-MB-231 breast cells to non-malignant MCF10A breast epithelial cells is reduced by DisBa-01 treatment. Inhibition of αvß3 integrin decreases CD63 expression in cancer cells suggesting an effect on SEV content. CONCLUSION: In summary, our findings demonstrate for the first time a key role of αvß3 integrin in cell-cell communication through SEVs. Video Abstract.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Extracellular Vesicles/metabolism , Integrin alphaVbeta3/metabolism , Breast/pathology , Cell Adhesion , Cell Line, Tumor , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Vesicles/ultrastructure , Female , Green Fluorescent Proteins/metabolism , Humans , Models, Biological , Protein Binding
9.
mSphere ; 5(4)2020 08 12.
Article in English | MEDLINE | ID: mdl-32817450

ABSTRACT

Extracellular vesicles (EVs) are membranous compartments produced by yeast and mycelial forms of several fungal species. One of the difficulties in perceiving the role of EVs during the fungal life, and particularly in cell wall biogenesis, is caused by the presence of a thick cell wall. One alternative to have better access to these vesicles is to use protoplasts. This approach has been investigated here with Aspergillus fumigatus, one of the most common opportunistic fungal pathogens worldwide. Analysis of regenerating protoplasts by scanning electron microscopy and fluorescence microscopy indicated the occurrence of outer membrane projections in association with surface components and the release of particles with properties resembling those of fungal EVs. EVs in culture supernatants were characterized by transmission electron microscopy and nanoparticle tracking analysis. Proteomic and glycome analysis of EVs revealed the presence of a complex array of enzymes related to lipid/sugar metabolism, pathogenic processes, and cell wall biosynthesis. Our data indicate that (i) EV production is a common feature of different morphological stages of this major fungal pathogen and (ii) protoplastic EVs are promising tools for undertaking studies of vesicle functions in fungal cells.IMPORTANCE Fungal cells use extracellular vesicles (EVs) to export biologically active molecules to the extracellular space. In this study, we used protoplasts of Aspergillus fumigatus, a major fungal pathogen, as a model to evaluate the role of EV production in cell wall biogenesis. Our results demonstrated that wall-less A. fumigatus exports plasma membrane-derived EVs containing a complex combination of proteins and glycans. Our report is the first to characterize fungal EVs in the absence of a cell wall. Our results suggest that protoplasts represent a promising model for functional studies of fungal vesicles.


Subject(s)
Aspergillus fumigatus/physiology , Extracellular Vesicles/physiology , Proteomics , Protoplasts/physiology , Cell Wall/metabolism , Extracellular Vesicles/ultrastructure , Fungal Proteins/metabolism , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Organelle Biogenesis , Protoplasts/ultrastructure
10.
Viruses ; 12(7)2020 07 16.
Article in English | MEDLINE | ID: mdl-32708544

ABSTRACT

Rotaviruses are the leading cause of viral gastroenteritis among children under five years of age. Rotavirus cell entry has been extensively studied; however, rotavirus cell release is still poorly understood. Specifically, the mechanism by which rotaviruses leave the cell before cell lysis is not known. Previous works have found rotavirus proteins and viral particles associated with extracellular vesicles secreted by cells. These vesicles have been shown to contain markers of exosomes; however, in a recent work they presented characteristics more typical of microparticles, and they were associated with an increase in the infectivity of the virus. In this work, we purified different types of vesicles from rotavirus-infected cells. We analyzed the association of virus with these vesicles and their possible role in promotion of rotavirus infection. We confirmed a non-lytic rotavirus release from the two cell lines tested, and observed a notable stimulation of vesicle secretion following rotavirus infection. A fraction of the secreted viral particles present in the cell supernatant was protected from protease treatment, possibly through its association with membranous vesicles; the more pronounced association of the virus was with fractions corresponding to cell membrane generated microvesicles. Using electron microscopy, we found different size vesicles with particles resembling rotaviruses associated from both- the outside and the inside. The viral particles inside the vesicles were refractory to neutralization with a potent rotavirus neutralizing monoclonal antibody, and were able to infect cells even without trypsin activation. The association of rotavirus particles with extracellular vesicles suggests these might have a role in virus spread.


Subject(s)
Extracellular Vesicles/virology , Rotavirus Infections/metabolism , Rotavirus/metabolism , Caco-2 Cells/virology , Extracellular Vesicles/ultrastructure , Humans , Microscopy, Electron, Transmission , Rotavirus/ultrastructure , Virion/metabolism , Virus Release
11.
Cells ; 9(4)2020 04 23.
Article in English | MEDLINE | ID: mdl-32340348

ABSTRACT

Mesenchymal stromal cells (MSCs) can generate immunological tolerance due to their regulatory activity in many immune cells. Extracellular vesicles (EVs) release is a pivotal mechanism by which MSCs exert their actions. In this study, we evaluate whether mesenchymal stromal cell extracellular vesicles (MSC-EVs) can modulate T cell response. MSCs were expanded and EVs were obtained by differential ultracentrifugation of the supernatant. The incorporation of MSC-EVs by T cells was detected by confocal microscopy. Expression of surface markers was detected by flow cytometry or CytoFLEX and cytokines were detected by RT-PCR, FACS and confocal microscopy and a miRNA PCR array was performed. We demonstrated that MSC-EVs were incorporated by lymphocytes in vitro and decreased T cell proliferation and Th1 differentiation. Interestingly, in Th1 polarization, MSC-EVs increased Foxp3 expression and generated a subpopulation of IFN-γ+/Foxp3+T cells with suppressive capacity. A differential expression profile of miRNAs in MSC-EVs-treated Th1 cells was seen, and also a modulation of one of their target genes, TGFbR2. MSC-EVs altered the metabolism of Th1-differentiated T cells, suggesting the involvement of the TGF-ß pathway in this metabolic modulation. The addition of MSC-EVs in vivo, in an OVA immunization model, generated cells Foxp3+. Thus, our findings suggest that MSC-EVs are able to specifically modulate activated T cells at an alternative regulatory profile by miRNAs and metabolism shifting.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Proliferation/genetics , Extracellular Vesicles/ultrastructure , Forkhead Transcription Factors/metabolism , Glycolysis , Membrane Potential, Mitochondrial , Mesenchymal Stem Cells/ultrastructure , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction/genetics , T-Lymphocytes, Regulatory/cytology
12.
Stem Cell Res Ther ; 10(1): 332, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31747944

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have been explored as promising tools for treatment of several neurological and neurodegenerative diseases. MSCs release abundant extracellular vesicles (EVs) containing a variety of biomolecules, including mRNAs, miRNAs, and proteins. We hypothesized that EVs derived from human Wharton's jelly would act as mediators of the communication between hMSCs and neurons and could protect hippocampal neurons from damage induced by Alzheimer's disease-linked amyloid beta oligomers (AßOs). METHODS: We isolated and characterized EVs released by human Wharton's jelly mesenchymal stem cells (hMSC-EVs). The neuroprotective action of hMSC-EVs was investigated in primary hippocampal cultures exposed to AßOs. RESULTS: hMSC-EVs were internalized by hippocampal cells in culture, and this was enhanced in the presence of AßOs in the medium. hMSC-EVs protected hippocampal neurons from oxidative stress and synapse damage induced by AßOs. Neuroprotection by hMSC-EVs was mediated by catalase and was abolished in the presence of the catalase inhibitor, aminotriazole. CONCLUSIONS: hMSC-EVs protected hippocampal neurons from damage induced by AßOs, and this was related to the transfer of enzymatically active catalase contained in EVs. Results suggest that hMSC-EVs should be further explored as a cell-free therapeutic approach to prevent neuronal damage in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/cytology , Neurons/pathology , Neuroprotection , Oxidative Stress , Synapses/pathology , Wharton Jelly/cytology , Animals , Biomarkers/metabolism , Catalase/metabolism , Exosomes/metabolism , Exosomes/ultrastructure , Extracellular Vesicles/drug effects , Extracellular Vesicles/ultrastructure , Hippocampus/pathology , Humans , Mesenchymal Stem Cells/drug effects , Neurons/drug effects , Neuroprotection/drug effects , Oxidative Stress/drug effects , Protein Multimerization , Rats , Reactive Oxygen Species/metabolism , Synapses/drug effects
13.
Int J Mol Sci ; 20(20)2019 Oct 09.
Article in English | MEDLINE | ID: mdl-31600881

ABSTRACT

Despite the different strategies used to treat ovarian cancer, around 70% of women/patients eventually fail to respond to the therapy. Cancer stem cells (CSCs) play a role in the treatment failure due to their chemoresistant properties. This capacity to resist chemotherapy allows CSCs to interact with different components of the tumor microenvironment, such as mesenchymal stem cells (MSCs), and thus contribute to tumorigenic processes. Although the participation of MSCs in tumor progression is well understood, it remains unclear how CSCs induce the pro-tumorigenic activity of MSCs in response to chemotherapy. Small extracellular vesicles, including exosomes, represent one possible way to modulate any type of cell. Therefore, in this study, we evaluate if small extracellular vesicle (sEV) derived from ovarian cancer spheroids (OCS), which are enriched in CSCs, can modify the activity of MSCs to a pro-tumorigenic phenotype. We show that sEV released by OCS in response to cisplatin induce an increase in the migration pattern of bone marrow MSCs (BM-MSCs) and the secretion interleukin-6 (IL-6), interleukin-8 (IL-8), and vascular endothelial growth factor A (VEGFA). Moreover, the factors secreted by BM-MSCs induce angiogenesis in endothelial cells and the migration of low-invasive ovarian cancer cells. These findings suggest that cisplatin could modulate the cargo of sEV released by CSCs, and these exosomes can further induce the pro-tumorigenic activity of MSCs.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cisplatin/pharmacology , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Ovarian Neoplasms/etiology , Ovarian Neoplasms/metabolism , Cell Line, Tumor , Cytokines/metabolism , Exosomes/metabolism , Exosomes/ultrastructure , Extracellular Vesicles/ultrastructure , Female , Gene Expression , Humans , Metalloproteases/genetics , Metalloproteases/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/pathology , Spheroids, Cellular , Tumor Microenvironment
14.
PLoS One ; 14(6): e0218616, 2019.
Article in English | MEDLINE | ID: mdl-31242249

ABSTRACT

AIM: To isolate and characterize oral extracellular vesicles from gingival crevicular fluid at 11-14 weeks and evaluate their capacity to identify patients at risk of developing gestational diabetes mellitus. METHODS: A case-control study was conducted, including patients who developed gestational diabetes mellitus (n = 11) and healthy pregnant controls (n = 23). Obstetric and periodontal histories were recorded at 11-14 weeks of gestation, and samples of gingival crevicular fluid obtained. Extracellular vesicles were isolated from gingival crevicular fluid by ExoQuick. Nanoparticle tracking analysis, ELISA and transmission electron microscopy were used to characterize extracellular vesicles. RESULTS: Total extracellular vesicles isolated from gingival crevicular fluid were significantly higher in patients who developed gestational diabetes mellitus later in pregnancy compared to normoglycemic pregnant women (6.3x109 vs 1.7 x1010, p value = 0.0026), and the concentration of the extracellular vesicles delivered an area under the ROC curve of 0.81. The distribution size of extracellular vesicles obtained using ExoQuick was around 148 ± 57 nm. There were no significant differences in the periodontal status between cases and controls. The exosome transmembrane protein CD63 was also detected in the extracellular vesicles of gingival crevicular fluid. CONCLUSION: We were able to isolate extracellular vesicles from gingival crevicular fluid using a method that is suitable to be applied in a clinical setting. Our results provide an insight into the potential capacity of first trimester oral extracellular vesicles as early biomarkers for the prediction of gestational diabetes mellitus in pre-symptomatic women.


Subject(s)
Diabetes, Gestational/etiology , Extracellular Vesicles/ultrastructure , Gingival Crevicular Fluid/cytology , Adult , Case-Control Studies , Diabetes, Gestational/diagnosis , Diabetes, Gestational/metabolism , Extracellular Vesicles/metabolism , Female , Gingival Crevicular Fluid/metabolism , Humans , Liquid Biopsy/methods , Particle Size , Periodontitis/complications , Periodontitis/metabolism , Periodontitis/pathology , Pregnancy , Pregnancy Trimester, First , Retrospective Studies , Risk Factors , Tetraspanin 30/metabolism
15.
mSphere ; 4(2)2019 03 20.
Article in English | MEDLINE | ID: mdl-30894430

ABSTRACT

Regular protocols for the isolation of fungal extracellular vesicles (EVs) are time-consuming, hard to reproduce, and produce low yields. In an attempt to improve the protocols used for EV isolation, we explored a model of vesicle production after growth of Cryptococcus gattii and Cryptococcus neoformans on solid media. Nanoparticle tracking analysis in combination with transmission electron microscopy revealed that C. gattii and C. neoformans produced EVs in solid media. The properties of cryptococcal vesicles varied according to the culture medium used and the EV-producing species. EV detection was reproduced with an acapsular mutant of C. neoformans, as well as with isolates of Candida albicans, Histoplasma capsulatum, and Saccharomyces cerevisiae Cryptococcal EVs produced in solid media were biologically active and contained regular vesicular components, including the major polysaccharide glucuronoxylomannan (GXM) and RNA. Since the protocol had higher yields and was much faster than the regular methods used for the isolation of fungal EVs, we asked if it would be applicable to address fundamental questions related to cryptococcal secretion. On the basis that polysaccharide export in Cryptococcus requires highly organized membrane traffic culminating with EV release, we analyzed the participation of a putative scramblase (Aim25; CNBG_3981) in EV-mediated GXM export and capsule formation in C. gattii EVs from a C. gattiiaim25Δ strain differed from those obtained from wild-type (WT) cells in physical-chemical properties and cargo. In a model of surface coating of an acapsular cryptococcal strain with vesicular GXM, EVs obtained from the aim25Δ mutant were more efficiently used as a source of capsular polysaccharides. Lack of the Aim25 scramblase resulted in disorganized membranes and increased capsular dimensions. These results associate the description of a novel protocol for the isolation of fungal EVs with the identification of a previously unknown regulator of polysaccharide release.IMPORTANCE Extracellular vesicles (EVs) are fundamental components of the physiology of cells from all kingdoms. In pathogenic fungi, they participate in important mechanisms of transfer of antifungal resistance and virulence, as well as in immune stimulation and prion transmission. However, studies on the functions of fungal EVs are still limited by the lack of efficient methods for isolation of these compartments. In this study, we developed an alternative protocol for isolation of fungal EVs and demonstrated an application of this new methodology in the study of the physiology of the fungal pathogen Cryptococcus gattii Our results describe a fast and reliable method for the study of fungal EVs and reveal the participation of scramblase, a phospholipid-translocating enzyme, in secretory processes of C. gattii.


Subject(s)
Cryptococcus gattii/enzymology , Extracellular Vesicles/chemistry , Fungal Polysaccharides/chemistry , Fungal Proteins/genetics , Mycology/methods , Biological Transport , Cryptococcus gattii/genetics , Cryptococcus neoformans/cytology , Cryptococcus neoformans/genetics , Extracellular Vesicles/ultrastructure , Microscopy, Electron, Transmission , Polysaccharides/genetics , Polysaccharides/isolation & purification
16.
Exp Mol Med ; 50(9): 1-12, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30201949

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) obtained from pluripotent stem cells (PSCs) constitute an interesting alternative to classical MSCs in regenerative medicine. Among their many mechanisms of action, MSC extracellular vesicles (EVs) are a potential suitable substitute for MSCs in future cell-free-based therapeutic approaches. Unlike cells, EVs do not elicit acute immune rejection, and they can be produced in large quantities and stored until ready to use. Although the therapeutic potential of MSC EVs has already been proven, a thorough characterization of MSC EVs is lacking. In this work, we used a label-free liquid chromatography tandem mass spectrometry proteomic approach to identify the most abundant proteins in EVs that are secreted from MSCs derived from PSCs (PD-MSCs) and from their parental induced PSCs (iPSCs). Next, we compared both datasets and found that while iPSC EVs enclose proteins that modulate RNA and microRNA stability and protein sorting, PD-MSC EVs are rich in proteins that organize extracellular matrix, regulate locomotion, and influence cell-substrate adhesion. Moreover, compared to their respective cells, iPSCs and iPSC EVs share a greater proportion of proteins, while the PD-MSC proteome appears to be more specific. Correlation and principal component analysis consistently aggregate iPSCs and iPSC EVs but segregate PD-MSC and their EVs. Altogether, these findings suggest that during differentiation, compared with their parental iPSC EVs, PD-MSC EVs acquire a more specific set of proteins; arguably, this difference might confer their therapeutic properties.


Subject(s)
Cell Differentiation , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proteomics , Cell Line , Extracellular Vesicles/ultrastructure , Humans , Principal Component Analysis , Stromal Cells/metabolism , Tandem Mass Spectrometry , Wharton Jelly/cytology
17.
Mem Inst Oswaldo Cruz ; 113(3): 178-184, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29412357

ABSTRACT

BACKGROUND: Members of the Bacteroides fragilis group are the most important components of the normal human gut microbiome, but are also major opportunistic pathogens that are responsible for significant mortality, especially in the case of bacteraemia and other severe infections, such as intra-abdominal abscesses. Up to now, several virulence factors have been described that might explain the involvement of B. fragilis in these infections. The secretion of extracellular membrane vesicles (EMVs) has been proposed to play a role in pathogenesis and symbiosis in gram-negative bacteria, by releasing soluble proteins and other molecules. In B. fragilis, these vesicles are known to have haemagglutination and sialidosis activities, and also contain a capsular polysaccharide (PSA), although their involvement in virulence is still not clear. OBJECTIVE: The aim of this study was to identify proteins in the EMV of the 638R B. fragilis strain by mass spectrometry, and also to assess for the presence of Bfp60, a surface plasminogen (Plg) activator, previously shown in B. fragilis to be responsible for the conversion of inactive Plg to active plasmin, which can also bind to laminin-1. METHODS: B. fragilis was cultured in a minimum defined media and EMVs were obtained by differential centrifugation, ultracentrifugation, and filtration. The purified EMVs were observed by both transmission electron microscopy (TEM) and immunoelectron microscopy (IM). To identify EMV constituent proteins, EMVs were separated by 1D SDS-PAGE and proteomic analysis of proteins sized 35 kDa to approximately 65 kDa was performed using mass spectrometry (MALDI-TOF MS). FINDINGS: TEM micrographs proved the presence of spherical vesicles and IM confirmed the presence of Bfp60 protein on their surface. Mass spectrometry identified 23 proteins with high confidence. One of the proteins from the B. fragilis EMVs was identified as an enolase P46 with a possible lyase activity. MAIN CONCLUSIONS: Although the Bfp60 protein was not detected by proteomics, α-enolase P46 was found to be present in the EMVs of B. fragilis. The P46 protein has been previously described to be present in the outer membrane of B. fragilis as an iron-regulated protein.


Subject(s)
Bacteroides fragilis/enzymology , Extracellular Vesicles/enzymology , Phosphopyruvate Hydratase/analysis , Bacteroides fragilis/ultrastructure , Electrophoresis, Polyacrylamide Gel , Extracellular Vesicles/ultrastructure , Humans , Laminin , Mass Spectrometry , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Phosphopyruvate Hydratase/metabolism , Plasminogen
18.
Stem Cell Rev Rep ; 14(2): 262-276, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29032399

ABSTRACT

Extracellular vesicles (EVs) released by mouse embryonic stem cells (mESCs) are considered a source of bioactive molecules that modulate their microenvironment by acting on intercellular communication. Either intracellular endosomal machinery or their derived EVs have been considered a relevant system of signal circuits processing. Herein, we show that these features are found in mESCs. Ultrastructural analysis revealed structures and organelles of the endosomal system such as coated pits and endocytosis-related vesicles, prominent rough endoplasmic reticulum and Golgi apparatus, and multivesicular bodies (MVBs) containing either few or many intraluminal vesicles (ILVs) that could be released as exosomes to extracellular milieu. Besides, budding vesicles shed from the plasma membrane to the extracellular space is suggestive of microvesicle biogenesis in mESCs. mESCs and mouse blastocyst express specific markers of the Endosomal Sorting Complex Required for Transport (ESCRT) system. Ultrastructural analysis and Nanoparticle Tracking Analysis (NTA) of isolated EVs revealed a heterogeneous population of exosomes and microvesicles released by mESCs. These vesicles contain Wnt10b and the Notch ligand Delta-like 4 (DLL4) and also the co-chaperone stress inducible protein 1 (STI1) and its partner Hsp90. Wnt10b and Dll4 colocalize with EVs biogenesis markers in mESCs. Overall, the present study supports the function of the mESCs endocytic network and their EVs as players in stem cell biology.


Subject(s)
Extracellular Vesicles/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Extracellular Vesicles/ultrastructure , Golgi Apparatus/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mouse Embryonic Stem Cells/ultrastructure , Multivesicular Bodies/metabolism
19.
Cell Mol Life Sci ; 75(12): 2211-2226, 2018 06.
Article in English | MEDLINE | ID: mdl-29222644

ABSTRACT

Trichomonas vaginalis is a common sexually transmitted parasite that colonizes the human urogenital tract, where it remains extracellular and adheres to epithelial cells. Infections range from asymptomatic to highly inflammatory, depending on the host and the parasite strain. Despite the serious consequences associated with trichomoniasis disease, little is known about parasite or host factors involved in attachment of the parasite-to-host epithelial cells. Here, we report the identification of microvesicle-like structures (MVs) released by T. vaginalis. MVs are considered universal transport vehicles for intercellular communication as they can incorporate peptides, proteins, lipids, miRNA, and mRNA, all of which can be transferred to target cells through receptor-ligand interactions, fusion with the cell membrane, and delivery of a functional cargo to the cytoplasm of the target cell. In the present study, we demonstrated that T. vaginalis release MVs from the plasma and the flagellar membranes of the parasite. We performed proteomic profiling of these structures demonstrating that they possess physical characteristics similar to mammalian extracellular vesicles and might be selectively charged with specific protein content. In addition, we demonstrated that viable T. vaginalis parasites release large vesicles (LVs), membrane structures larger than 1 µm that are able to interact with other parasites and with the host cell. Finally, we show that both populations of vesicles present on the surface of T vaginalis are induced in the presence of host cells, consistent with a role in modulating cell interactions.


Subject(s)
Extracellular Vesicles/metabolism , Host-Parasite Interactions , Trichomonas Vaginitis/metabolism , Trichomonas Vaginitis/parasitology , Trichomonas vaginalis/physiology , Trichomonas vaginalis/ultrastructure , Cell Communication , Extracellular Vesicles/chemistry , Extracellular Vesicles/ultrastructure , Female , HeLa Cells , Humans , Proteomics , Protozoan Proteins/analysis , Protozoan Proteins/metabolism , Trichomonas vaginalis/chemistry , Trichomonas vaginalis/cytology
20.
Mol Carcinog ; 57(4): 503-511, 2018 04.
Article in English | MEDLINE | ID: mdl-29247548

ABSTRACT

There is increasing evidence that cancer dissemination and metastasis establishment may not only be due to the movement of tumor cells. Content of extracellular vesicles (EVs) secreted by tumor cells may also reflect the origin of these cells. Some molecules that constitute these EVs have already been used as targets for detection of specific tumors. However, to the best of our knowledge, EVs from biopsies and plasma have not yet been compared nor thoroughly investigated as triggers of malignant transformation and metastatic niche formation. To evaluate the role of EVs in the cellular microenvironment, we have treated the normal epithelial prostate cell lines, RWPE-1 and PNT-2, with a pool of EVs from biopsies of prostate primary tumors (bEVs), biopsies of benign prostate hyperplasia (hEVs), plasma of prostate cancer (PCa) patients (pEVs) or plasma of healthy individuals (pnEVs). Each of the four pools consisted of isolated EVs from several subjects, of which PCa patients were in different stages of cancer. Migration and proliferation profiles, cytokine release, and a panel of PCa-associated genes' expression of epithelial-mesenchymal transition in the cell lines were evaluated after 24 h incubation with EVs. When compared to the control groups, cells treated with the pool of EVs isolated from tumor biopsies and plasma of PCa patients showed greater migration and proliferation, significant alterations in gene expression, and high levels of IL-8, factors that are associated with cancer development. Specifically, isolated bEVs and pEVs may induce malignant features in non-tumor cells by activating several cellular events associated with cancer progression, suggesting that future PCa therapy may target multiple elements found in tumor-derived EVs.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Extracellular Vesicles/genetics , Prostate/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line , Cell Movement/genetics , Extracellular Vesicles/ultrastructure , Gene Expression Regulation, Neoplastic , Humans , Male , Microscopy, Electron, Scanning , Middle Aged , Prostate/cytology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tumor Cells, Cultured , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL