Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Microb Biotechnol ; 16(7): 1524-1535, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37212362

RESUMO

Exosomes, membrane vesicles released extracellularly from cells, contain nucleic acids, proteins, lipids and other components, allowing the transfer of material information between cells. Recent studies reported the role of exosomes in pathogenic microbial infection and host immune mechanisms. Brucella-invasive bodies can survive in host cells for a long time and cause chronic infection, which causes tissue damage. Whether exosomes are involved in host anti-Brucella congenital immune responses has not been reported. Here, we extracted and identified exosomes secreted by Brucella melitensis M5 (Exo-M5)-infected macrophages, and performed in vivo and in vitro studies to examine the effects of exosomes carrying antigen on the polarization of macrophages and immune activation. Exo-M5 promoted the polarization of M1 macrophages, which induced the significant secretion of M1 cytokines (tumour necrosis factor-α and interferon-γ) through NF-κB signalling pathways and inhibited the secretion of M2 cytokines (IL-10), thereby inhibiting the intracellular survival of Brucella. Exo-M5 activated innate immunity and promoted the release of IgG2a antibodies that protected mice from Brucella infection and reduced the parasitaemia of Brucella in the spleen. Furthermore, Exo-M5 contained Brucella antigen components, including Omp31 and OmpA. These results demonstrated that exosomes have an important role in immune responses against Brucella, which might help elucidate the mechanisms of host immunity against Brucella infection and aid the search for Brucella biomarkers and the development of new vaccine candidates.


Assuntos
Brucelose , Exossomos , Macrófagos , Brucella melitensis , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/microbiologia , Exossomos/imunologia , Exossomos/microbiologia , Animais , Camundongos , Polaridade Celular , Antígenos de Bactérias/imunologia , Brucelose/imunologia , Brucelose/metabolismo , Transdução de Sinais , Espaço Intracelular/microbiologia , Viabilidade Microbiana
2.
Nutrients ; 14(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35057528

RESUMO

Gut bacteria release extracellular vesicles (BEVs) as an intercellular communication mechanism that primes the host innate immune system. BEVs from E. coli activate dendritic cells (DCs) and subsequent T-cell responses in a strain-specific manner. The specific immunomodulatory effects were, in part, mediated by differential regulation of miRNAs. This study aimed to deepen understanding of the mechanisms of BEVs to drive specific immune responses by analyzing their impact on DC-secreted cytokines and exosomes. DCs were challenged with BEVs from probiotic and commensal E. coli strains. The ability of DC-secreted factors to activate T-cell responses was assessed by cytokine quantification in indirect DCs/naïve CD4+ T-cells co-cultures on Transwell supports. DC-exosomes were characterized in terms of costimulatory molecules and miRNAs cargo. In the absence of direct cellular contacts, DC-secreted factors triggered secretion of effector cytokines by T-cells with the same trend as direct DC/T-cell co-cultures. The main differences between the strains influenced the production of Th1- and Treg-specific cytokines. Exosomes released by BEV-activated DCs were enriched in surface proteins involved in antigen presentation and T-cell activation, but differed in the content of immune-related miRNA, depending on the origin of the BEVs. These differences were consistent with the derived immune responses.


Assuntos
Citocinas/metabolismo , Células Dendríticas/microbiologia , Exossomos/microbiologia , Vesículas Extracelulares/imunologia , Microbioma Gastrointestinal/imunologia , Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Comunicação Celular/imunologia , Técnicas de Cocultura , Escherichia coli/imunologia , Exossomos/imunologia , Humanos , Ativação Linfocitária/imunologia , MicroRNAs/metabolismo , Probióticos/administração & dosagem , Linfócitos T/imunologia , Linfócitos T/microbiologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/microbiologia
3.
Oxid Med Cell Longev ; 2021: 8460355, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34367468

RESUMO

Mastitis is a common disease in the dairy industry that causes huge economic losses worldwide. Exosomes (carrying proteins, miRNA, lncRNA, etc.) play a vital role in the regulation of immune response. lncRNA can play a variety of regulatory roles by combining with protein, RNA, and DNA. The expression of mRNA and lncRNA in exosomes derived from bovine mammary epithelial cells infected by S. aureus is rarely understood. To explore this issue, RNA sequencing analysis was performed on exosomes derived from S. aureus-infected and noninfected MAC-T cells. Analysis of the sequencing results showed that there were 186 differentially expressed genes, 431 differentially expressed mRNAs and 19 differentially expressed lncRNAs in the exosomes derived from S. aureus-infected and noninfected MAC-T cells. By predicting lncRNA target genes, it was found that 19 differentially expressed lncRNAs all acted on multiple mRNAs in cis and trans. GO analysis revealed that differentially expressed genes and lncRNA target genes played significant roles in such metabolism (reactive oxygen species metabolic processes), transmembrane transport, cellular response to DNA damage stimulus, and response to cytokines. KEGG enrichment indicated that lncRNA target genes gathered in the TNF pathway, Notch pathway, MAPK pathway, NF-kappa B pathway, Hippo pathway, p53 pathway, reactive oxygen species metabolic processes, and longevity regulating pathway. In summary, all data indicated that differentially expressed gene, mRNA, and lncRNA in transcriptional profiling of exosomes participated in bacterial invasion and adhesion, oxidative stress, inflammation, and apoptosis-related signaling pathway. The data obtained in this study would provide valuable resource for understanding the lncRNA information in exosomes derived from dairy cow mammary epithelial cells and conduced to the study of S. aureus infection in dairy cow mammary glands.


Assuntos
Células Epiteliais/metabolismo , Exossomos/genética , Glândulas Mamárias Animais/metabolismo , Mastite Bovina/patologia , Infecções Estafilocócicas/genética , Staphylococcus aureus/fisiologia , Transcriptoma , Animais , Bovinos , Células Epiteliais/microbiologia , Exossomos/metabolismo , Exossomos/microbiologia , Feminino , Glândulas Mamárias Animais/microbiologia , Mastite Bovina/genética , Mastite Bovina/metabolismo , Mastite Bovina/microbiologia , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA-Seq , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/veterinária
4.
Gastroenterology ; 161(5): 1552-1566.e12, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34371001

RESUMO

BACKGROUND & AIMS: Enterotoxigenic Bacteroides fragilis (ETBF) is strongly associated with the occurrence of inflammatory bowel disease (IBD), colitis-associated colorectal cancer, and colorectal cancer (CRC). However, the mechanism of ETBF-induced intestinal inflammation and tumorigenesis remains unclear. METHODS: microRNA sequencing was used to detect the differentially expressed microRNAs in both ETBF-treated cells and exosomes derived from ETBF-inoculated cells. Cell Counting Kit 8 assays were used to evaluate the effect of ETBF and exosomes on CRC cell proliferation. The biological role and mechanism of ETBF-mediated miR-149-3p in colitis and colon carcinogenesis were determined both in vitro and in vivo. RESULTS: ETBF promoted CRC cell proliferation by down-regulating miR-149-3p both in vitro and in vivo. ETBF-down-regulated miR-149-3p depended on METTL14-mediated N6-methyladenosine methylation. As the target gene of miR-149-3p, PHF5A transactivated SOD2 through regulating KAT2A messenger RNA alternative splicing after ETBF treatment in CRC cells. miR-149-3p could be released in exosomes and mediated intercellular communication by modulating T-helper type 17 cell differentiation. The level of plasma exosomal miR-149-3p was gradually decreased from healthy control individuals to patients with IBD and CRC. miR-149-3p, existing in plasma exosomes, negatively correlated with the abundance of ETBF in patients with IBD and CRC. CONCLUSIONS: Exosomal miR-149-3p derived from ETBF-treated cells facilitated T-helper type 17 cell differentiation. ETBF-induced colorectal carcinogenesis depended on down-regulating miR-149-3p and further promoting PHF5A-mediated RNA alternative splicing of KAT2A in CRC cells. Targeting the ETBF/miR-149-3p pathway presents a promising approach to treat patients with intestinal inflammation and CRC with a high amount of ETBF.


Assuntos
Bacteroides fragilis/patogenicidade , Colite Ulcerativa/microbiologia , Colo/microbiologia , Neoplasias Colorretais/microbiologia , Doença de Crohn/microbiologia , Exossomos/microbiologia , MicroRNAs/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Colite Ulcerativa/genética , Colite Ulcerativa/metabolismo , Colite Ulcerativa/patologia , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Doença de Crohn/genética , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Modelos Animais de Doenças , Exossomos/genética , Exossomos/metabolismo , Células HCT116 , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , MicroRNAs/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Transativadores/genética , Transativadores/metabolismo
5.
Front Immunol ; 12: 628973, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33868247

RESUMO

Exosomes are cell-derived nanovesicles carrying protein, lipid, and nucleic acid for secreting cells, and act as significant signal transport vectors for cell-cell communication and immune modulation. Immune-cell-derived exosomes have been found to contain molecules involved in immunological pathways, such as MHCII, cytokines, and pathogenic antigens. Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), remains one of the most fatal infectious diseases. The pathogen for tuberculosis escapes the immune defense and continues to replicate despite rigorous and complicate host cell mechanisms. The infected-cell-derived exosomes under this circumstance are found to trigger different immune responses, such as inflammation, antigen presentation, and activate subsequent pathways, highlighting the critical role of exosomes in anti-MTB immune response. Additionally, as a novel kind of delivery system, exosomes show potential in developing new vaccination and treatment of tuberculosis. We here summarize recent research progress regarding exosomes in the immune environment during MTB infection, and further discuss the potential of exosomes as delivery system for novel anti-MTB vaccines and therapies.


Assuntos
Exossomos/transplante , Imunoterapia , Mycobacterium tuberculosis/patogenicidade , Vacinas contra a Tuberculose/uso terapêutico , Tuberculose/terapia , Imunidade Adaptativa , Animais , Autofagia , Exossomos/imunologia , Exossomos/microbiologia , Humanos , Evasão da Resposta Imune , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Tuberculose/microbiologia
6.
Eur J Clin Microbiol Infect Dis ; 40(11): 2427-2430, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33893878

RESUMO

Exosomes are selectively packaged cell-derived vesicles that contain a rich cargo of nucleic acids and proteins. The small heat shock protein, Hsp16.3, is an important capsule protein produced by Mycobacterium tuberculosis (MTB). Exploring the distribution of Hsp16.3 in exosomes is valuable to tuberculosis biomarker development. Our results showed that Hsp16.3 protein overexpressed in cells can be efficiently packaged into exosomes. U937 cells infected with MTB secreted abnormally excessive amounts of Hsp16.3 protein in exosomes. Finally, a substantial number of Hsp16.3 proteins were detected in blood exosomes of tuberculosis patients. The research provides a potential exosome-based tuberculosis biomarker for MTB diagnosis.


Assuntos
Proteínas de Bactérias/metabolismo , Biomarcadores/análise , Chaperoninas/metabolismo , Exossomos/metabolismo , Mycobacterium tuberculosis/metabolismo , Tuberculose/microbiologia , Proteínas de Bactérias/genética , Biomarcadores/metabolismo , Linhagem Celular , Chaperoninas/genética , Exossomos/genética , Exossomos/microbiologia , Humanos , Mycobacterium tuberculosis/genética , Tuberculose/diagnóstico , Tuberculose/genética , Tuberculose/metabolismo
7.
Curr Opin Insect Sci ; 40: 39-47, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32590312

RESUMO

Ticks and mosquitoes are medically important vectors that transmit several pathogens, including arboviruses, to humans. Understanding how these blood-feeding arthropods transmit pathogens to humans requires knowledge on the molecular and cellular interplay at vector-host interface. Recent studies have highlighted the role of tick and mosquito small extracellular vesicles (EVs), including exosomes, facilitating arbovirus transmission within arthropod cells and from arthropod to mammalian cells. In this review, we summarize this emerging line of investigation in understanding the role of tick and mosquito exosomes in vector-pathogen-host tripartite interactions. Understanding the role of arthropod exosomes in pathogen interactions could lead to the discovery of novel therapeutic targets to interfere with the life cycle of several pathogens transmitted by vectors.


Assuntos
Arbovírus/fisiologia , Artrópodes/fisiologia , Exossomos/fisiologia , Doenças Transmitidas por Vetores/transmissão , Animais , Exossomos/microbiologia , Exossomos/parasitologia , Exossomos/virologia , Doenças Transmitidas por Vetores/microbiologia , Doenças Transmitidas por Vetores/parasitologia , Doenças Transmitidas por Vetores/virologia
8.
Gut Microbes ; 11(6): 1677-1694, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32583714

RESUMO

Adherent-invasive E. coli (AIEC), which abnormally colonize the intestinal mucosa of Crohn's disease (CD) patients, are able to adhere to and invade intestinal epithelial cells (IECs), survive and replicate within macrophages and induce a pro-inflammatory response. AIEC infection of IECs induces secretion of exosomes that increase AIEC replication in exosome-receiving IECs and macrophages. Here, we investigated the mechanism underlying the increased AIEC replication in cells receiving exosomes from AIEC-infected cells. Exosomes released by uninfected human intestinal epithelial T84 cells (Exo-uninfected) or by T84 cells infected with the clinical AIEC LF82 strain (Exo-LF82), the nonpathogenic E. coli K12 strain (Exo-K12) or the commensal E. coli HS strain (Exo-HS) were purified and used to stimulate T84 cells. Stimulation of T84 cells with Exo-LF82 inhibited autophagy compared with Exo-uninfected, Exo-K12 and Exo-HS. qRT-PCR analysis revealed increased levels of miR-30c and miR-130a in Exo-LF82 compared to Exo-uninfected, Exo-K12 and Exo-HS. These miRNAs were transferred via exosomes to recipient cells, in which they targeted and inhibited ATG5 and ATG16L1 expression and thereby autophagy response, thus favoring AIEC intracellular replication. Inhibition of these miRNAs in exosome-donor cells infected with AIEC LF82 abolished the increase in miR-30c and miR-130a levels in the released Exo-LF82 and in Exo-LF82-receiving cells, thus suppressing the inhibitory effect of Exo-LF82 on ATG5 and ATG16L1 expression and on autophagy-mediated AIEC clearance in Exo-LF82-receiving cells. Our study shows that upon AIEC infection, IECs secrete exosomes that can transfer specific miRNAs to recipient IECs, inhibiting autophagy-mediated clearance of intracellular AIEC.


Assuntos
Autofagia , Doença de Crohn/microbiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli/fisiologia , Exossomos/microbiologia , MicroRNAs/metabolismo , Animais , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Aderência Bacteriana , Transporte Biológico , Linhagem Celular , Doença de Crohn/genética , Doença de Crohn/metabolismo , Doença de Crohn/fisiopatologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/citologia , Escherichia coli/genética , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/fisiopatologia , Exossomos/genética , Exossomos/metabolismo , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , MicroRNAs/genética
9.
J Am Heart Assoc ; 9(6): e014120, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32174233

RESUMO

Background Epidemiological studies have suggested an association between Helicobacter pylori (H pylori) infection and atherosclerosis through undefined mechanisms. Endothelial dysfunction is critical to the development of atherosclerosis and related cardiovascular diseases. The present study was designed to test the hypothesis that H pylori infection impaires endothelial function through exosome-mediated mechanisms. Methods and Results Young male and female patients (18-35 years old) with and without H pylori infection were recruited to minimize the chance of potential risk factors for endothelial dysfunction for the study. Endothelium-dependent flow-mediated vasodilatation of the brachial artery was evaluated in the patients and control subjects. Mouse infection models with CagA+H pylori from a gastric ulcer patient were created to determine if H pylori infection-induced endothelial dysfunction could be reproduced in animal models. H pylori infection significantly decreased endothelium-dependent flow-mediated vasodilatation in young patients and significantly attenuated acetylcholine-induced endothelium-dependent aortic relaxation without change in nitroglycerin-induced endothelium-independent vascular relaxation in mice. H pylori eradication significantly improved endothelium-dependent vasodilation in both patients and mice with H pylori infection. Exosomes from conditioned media of human gastric epithelial cells cultured with CagA+H pylori or serum exosomes from patients and mice with H pylori infection significantly decreased endothelial functions with decreased migration, tube formation, and proliferation in vitro. Inhibition of exosome secretion with GW4869 effectively preserved endothelial function in mice with H pylori infection. Conclusions H pylori infection impaired endothelial function in patients and mice through exosome-medicated mechanisms. The findings indicated that H pylori infection might be a novel risk factor for cardiovascular diseases.


Assuntos
Artéria Braquial/microbiologia , Células Endoteliais/microbiologia , Endotélio Vascular/microbiologia , Exossomos/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Vasodilatação , Adolescente , Adulto , Compostos de Anilina/farmacologia , Animais , Antibacterianos/uso terapêutico , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Compostos de Benzilideno/farmacologia , Artéria Braquial/metabolismo , Artéria Braquial/fisiopatologia , Estudos de Casos e Controles , Linhagem Celular , Movimento Celular , Proliferação de Células , China , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Exossomos/efeitos dos fármacos , Exossomos/metabolismo , Feminino , Fármacos Gastrointestinais/uso terapêutico , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/fisiopatologia , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Masculino , Camundongos Endogâmicos C57BL , Missouri , Neovascularização Fisiológica , Vasodilatação/efeitos dos fármacos , Adulto Jovem
10.
Mol Cell Endocrinol ; 506: 110756, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32045626

RESUMO

Exosomes have been intensively studied in autoimmune diseases, and circulating exosomes and microvesicles have also been explored in autoimmune thyroiditis (AITD). However, the role of thyroid cell-derived exosomes in immune responses is unclear. We showed that IFN-γ-treated Nthy-ori 3-1 cell-derived exosomes (IFN-γ-Exo) harbored TPO, HSP60 and MHC-II and activated dendritic cells (DCs) in vitro. Compared with Exo-targeted DCs (DCExo), IFN-γ-Exo-targeted DCs (DCIFN-γ-Exo) promoted the expression and release of proinflammatory cytokines, such as IFN-γ, IL-17A and IL-22, from CD4+ T lymphocytes and inhibited the expression and release of anti-inflammatory cytokines, such as IL-4, IL-10 and TGF-ß1; however, IFN-γ-Exo did not have this effect compared with Nthy-ori 3-1 cell-derived exosomes (Exo). DCIFN-γ-Exo stimulates the expression and release of cytokines from CD4+ T lymphocytes more efficiently than IFN-γ-Exo. Thus, DCIFN-γ-Exo may effectively induce CD4+ T lymphocyte-mediated immune responses and play a role in the occurrence and development of AITD.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Exossomos/metabolismo , Exossomos/microbiologia , Células Epiteliais da Tireoide/metabolismo , Adulto , Autoantígenos/metabolismo , Células Cultivadas , Chaperonina 60/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Exossomos/efeitos dos fármacos , Feminino , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Interferon gama/farmacologia , Iodeto Peroxidase/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Pessoa de Meia-Idade , Proteínas Mitocondriais/metabolismo , Células Epiteliais da Tireoide/efeitos dos fármacos
11.
Int J Chron Obstruct Pulmon Dis ; 14: 2563-2573, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31819394

RESUMO

Background: Pseudomonas aeruginosa (PA) colonization confers poor prognosis in bronchiectasis. However, the biomarkers and biological pathways underlying these associations are unclear. Objective: To identify the roles of PA colonization in bronchiectasis by exploring for sputum exosomal microRNA profiles. Methods: We enrolled 98 patients with clinically stable bronchiectasis and 17 healthy subjects. Sputum was split for bacterial culture and exosomal microRNA sequencing, followed by validation with quantitative polymerase chain reaction. Bronchiectasis patients were stratified into PA and non-PA colonization groups based on sputum culture findings. We applied Gene Ontology and Kyoto Encyclopedia of Genes and Genome pathway enrichment analysis to explore biological pathways corresponding to the differentially expressed microRNAs (DEMs) associated with PA colonization. Results: Eighty-two bronchiectasis patients and 9 healthy subjects yielded sufficient sputum that passed quality control. We identified 10 overlap DEMs for the comparison between bronchiectasis patients and healthy subjects, and between PA and non-PA colonization group. Both miR-92b-5p and miR-223-3p could discriminate PA colonization (C-statistic >0.60) and independently correlated with PA colonization in multiple linear regression analysis. The differential expression of miR-92b-5p was validated by quantitative polymerase chain reaction (P<0.05), whereas the differential expression of miR-223 trended towards statistical significance (P=0.06). These DEMs, whose expression levels correlated significantly with sputum inflammatory biomarkers (interleukin-1ß and interleukin-8) level, were implicated in the modulation of the nuclear factor-κB, phosphatidylinositol and longevity regulation pathways. Conclusion: Sputum exosomal microRNAs are implicated in PA colonization in bronchiectasis, highlighting candidate targets for therapeutic interventions to mitigate the adverse impacts conferred by PA colonization.


Assuntos
Bronquiectasia/genética , Bronquiectasia/microbiologia , Exossomos/genética , Exossomos/microbiologia , MicroRNAs/genética , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/crescimento & desenvolvimento , Adulto , Bronquiectasia/diagnóstico , Estudos de Casos e Controles , Feminino , Redes Reguladoras de Genes , Interações Hospedeiro-Patógeno , Humanos , Masculino , Pessoa de Meia-Idade , Infecções por Pseudomonas/diagnóstico , Transdução de Sinais , Escarro/química , Escarro/microbiologia
12.
J Cell Sci ; 132(6)2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30886004

RESUMO

Vector-borne diseases cause over 700,000 deaths annually and represent 17% of all infectious illnesses worldwide. This public health menace highlights the importance of understanding how arthropod vectors, microbes and their mammalian hosts interact. Currently, an emphasis of the scientific enterprise is at the vector-host interface where human pathogens are acquired and transmitted. At this spatial junction, arthropod effector molecules are secreted, enabling microbial pathogenesis and disease. Extracellular vesicles manipulate signaling networks by carrying proteins, lipids, carbohydrates and regulatory nucleic acids. Therefore, they are well positioned to aid in cell-to-cell communication and mediate molecular interactions. This Review briefly discusses exosome and microvesicle biogenesis, their cargo, and the role that nanovesicles play during pathogen spread, host colonization and disease pathogenesis. We then focus on the role of extracellular vesicles in dictating microbial pathogenesis and host immunity during transmission of vector-borne pathogens.


Assuntos
Vetores Artrópodes , Vesículas Extracelulares , Doenças Transmitidas por Vetores , Amebíase/parasitologia , Amebíase/transmissão , Animais , Vetores Artrópodes/microbiologia , Vetores Artrópodes/parasitologia , Culicidae/microbiologia , Culicidae/parasitologia , Vetores de Doenças , Exossomos/imunologia , Exossomos/microbiologia , Exossomos/parasitologia , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/microbiologia , Vesículas Extracelulares/parasitologia , Filariose/parasitologia , Filariose/transmissão , Hemípteros/microbiologia , Hemípteros/parasitologia , Interações Hospedeiro-Parasita/imunologia , Interações Hospedeiro-Parasita/fisiologia , Humanos , Imunomodulação , Leishmaniose/parasitologia , Leishmaniose/transmissão , Malária/parasitologia , Malária/transmissão , Psychodidae/microbiologia , Psychodidae/parasitologia , Tripanossomíase/parasitologia , Tripanossomíase/transmissão , Doenças Transmitidas por Vetores/microbiologia , Doenças Transmitidas por Vetores/parasitologia , Doenças Transmitidas por Vetores/transmissão , Viroses/microbiologia , Viroses/transmissão
13.
Cell Microbiol ; 21(7): e13020, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30817089

RESUMO

Unlike urinary tract infection (UTI), asymptomatic bacteriuria (ABU) should not be treated, with some exceptions such as pregnant women and patients who will undergo traumatic urologic interventions. However, there has been no clinically available marker for their differential diagnosis. Exosomes or small extracellular vesicles carry proteins contained in cells from which they are derived, thus having the potential as a biomarker of several diseases. On the basis of the hypothesis that the molecular signature of exosomes in urine may differ between UTI and ABU patients, we examined if urinary exosomes could serve as a marker for their differential diagnosis. Exosomes were isolated by ultracentrifugation or affinity-based method from cell culture medium of monocytic THP-1 and uroepithelial SV-HUC-1 cells and human urine. Protein expression was examined by Western blot analysis, ELISA, and CLEIA. The results showed that the levels of intracellular signalling molecules Akt and ERK and transcription factor NF-κB increased in exosomes isolated from THP-1 and SV-HUC-1 cells cocultured with Escherichia coli and/or treated with lipopolysaccharide. In urinary exosomes of UTI patients, Akt significantly diminished, and an exosomal marker CD9 showed a trend to decrease after treatment with antimicrobial agents. More importantly, Akt and CD9 levels in urinary exosomes were higher in UTI patients than in ABU patients, which was also observed after correction by urine creatinine. Collectively, these results suggest that Akt and CD9 in urinary exosomes could be useful markers for differential diagnosis of UTI and ABU.


Assuntos
Bacteriúria/urina , Exossomos/genética , Proteínas Proto-Oncogênicas c-akt/urina , Tetraspanina 29/urina , Infecções Urinárias/urina , Bacteriúria/microbiologia , Bacteriúria/patologia , Biomarcadores/urina , Diagnóstico Diferencial , Escherichia coli/genética , Exossomos/microbiologia , Feminino , Regulação da Expressão Gênica/genética , Humanos , Lipopolissacarídeos/farmacologia , Monócitos/patologia , Gravidez , Infecções Urinárias/genética , Infecções Urinárias/microbiologia
14.
Clin Exp Immunol ; 194(3): 339-349, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30105789

RESUMO

Emerging evidence has linked the exosomes to many immunological disorders, including infectious diseases. However, knowledge regarding the role of exosomes in Helicobacter pylori infection is limited. Here, we show that serum exosomes from chronic gastritis patients with H. pylori infection (Hp exosomes) stimulate the expression of the soluble interleukin (IL)-6 receptor (sIL-6R), which is involved in IL-6 trans-signalling in gastric epithelial cells. Interestingly, sIL-6R up-regulates expression of the proinflammatory cytokine IL-1α, and the neutralization of sIL-6R suppresses IL-1α secretion. Thus, Hp exosomes regulate IL-1α expression via sIL-6R-mediated IL-6 trans-signaling. Altogether, this study reveals a novel perspective in which exosomes play a vital role in immunological mechanisms during H. pylori infection.


Assuntos
Células Epiteliais/metabolismo , Exossomos/microbiologia , Mucosa Gástrica/metabolismo , Gastrite/microbiologia , Helicobacter pylori/imunologia , Interleucina-1alfa/biossíntese , Interleucina-6/imunologia , Receptores de Interleucina-6/metabolismo , Células Cultivadas , Criança , Feminino , Mucosa Gástrica/citologia , Gastrite/imunologia , Infecções por Helicobacter/imunologia , Humanos , Masculino , Receptores de Interleucina-6/biossíntese
15.
J Cell Mol Med ; 22(11): 5708-5719, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30160350

RESUMO

Helicobacter pylori (H. pylori) infection triggers chronic inflammation that has been associated with gastric cancer (GC). Exosomes are small extracellular vesicles that have become the key mediators of intercellular communication. In this study, we investigated exosome-mediated communication between H. pylori-infected GC cells and macrophages, focusing on the transfer of activated mesenchymal-epithelial transition factor (MET). We observed a significant decrease in MET protein expression in GC cells after infection with H. pylori, whereas MET mRNA levels remained unchanged. Intriguingly, MET expression, specifically the phosphorylated active form, was increased in exosomes released from H. pylori-infected GC cells. Confocal microscopy and Western blotting analyses showed that these exosomes containing MET were delivered to and internalized by macrophages. Indeed, in human GC tissues positive for H. pylori, we also observed that activated MET was highly expressed in tumour-infiltrating macrophages. After internalization, exosomal MET then appeared to educate the macrophages towards a pro-tumorigenesis phenotype. This included exosomal MET-mediated stimulation of proinflammatory cytokine secretion IL-1ß, which subsequently promoted tumour growth and progression in vitro and in vivo. Taken together, these data were the first to demonstrate H. pylori infection-induced upregulation of activated MET in exosomes and the pro-tumorigenic effect on tumour-associated macrophages.


Assuntos
Infecções por Helicobacter/genética , Inflamação/genética , Proteínas Proto-Oncogênicas c-met/genética , Neoplasias Gástricas/genética , Animais , Linhagem Celular Tumoral , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Exossomos/genética , Exossomos/microbiologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/complicações , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Xenoenxertos , Humanos , Inflamação/microbiologia , Inflamação/patologia , Interleucina-1beta/genética , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Neoplasias Gástricas/complicações , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia
16.
Cell Stress Chaperones ; 23(4): 663-672, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29383581

RESUMO

Bovine milk is rich in exosomes, which contain abundant miRNAs and play important roles in the regulation of neonatal growth and development of adaptive immunity. Here, we analyzed miRNA expression profiles of bovine milk exosomes from three healthy and three mastitic cows, and then six miRNA libraries were constructed. Interestingly, we detected no scRNAs and few snRNAs in milk exosomes; this result indicated a potential preference for RNA packaging in milk exosomes. A total of 492 known and 980 novel exosomal miRNAs were detected, and the 10 most expressed miRNAs in the six samples accounted for 80-90% of total miRNA-associated reads. Expression analyses identified 18 miRNAs with significantly different expression between healthy and infected animals; the predicted target genes of differentially expressed miRNAs were significantly enriched in immune system process, response to stimulus, growth, etc. Moreover, target genes were significantly enriched in several Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways including inflammatory, immune, and cancer pathways. Our survey provided comprehensive information about milk exosomes and exosomal miRNAs involved in mastitis. Moreover, the differentially expressed miRNAs, especially miR-223 and miR-142-5p, could be considered as potential candidates for mastitis.


Assuntos
Bovinos/genética , Exossomos/genética , Exossomos/microbiologia , Perfilação da Expressão Gênica , Genoma , MicroRNAs/genética , Leite/metabolismo , Staphylococcus aureus/fisiologia , Animais , Exossomos/ultraestrutura , Feminino , Regulação da Expressão Gênica , Ontologia Genética , MicroRNAs/metabolismo , Reprodutibilidade dos Testes , Análise de Sequência de RNA
17.
FASEB J ; 32(1): 97-110, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28855278

RESUMO

Acute lung injury (ALI) is a common cause of morbidity in patients after severe injury due to dysregulated inflammation, which is believed to be driven by gut-derived inflammatory mediators carried via mesenteric lymph (ML). We have previously demonstrated that nano-sized extracellular vesicles, called exosomes, secreted into ML after trauma/hemorrhagic shock (T/HS) have the potential to activate immune cells in vitro Here, we assess the function of ML exosomes in the development of T/HS-induced ALI and the role of TLR4 in the ML exosome-mediated inflammatory response. ML exosomes isolated from rats subjected to T/HS stimulated NF-κB activation and caused proinflammatory cytokine production in alveolar macrophages. In vivo experiments revealed that intravenous injection of exosomes harvested after T/HS, but not before shock, caused recruitment of inflammatory cells in the lung, increased vascular permeability, and induced histologic ALI in naive mice. The exosome-depleted supernatant of ML had no effect on in vitro and in vivo inflammatory responses. We also demonstrated that both pharmacologic inhibition and genetic knockout of TLR4 completely abolished ML exosome-induced cytokine production in macrophages. Thus, our findings define the critical role of exosomes secreted into ML as a critical mediator of T/HS-induced ALI through macrophage TLR4 activation.-Kojima, M., Gimenes-Junior, J. A., Chan, T. W., Eliceiri, B. P., Baird, A., Costantini, T. W., Coimbra, R. Exosomes in postshock mesenteric lymph are key mediators of acute lung injury triggering the macrophage activation via Toll-like receptor 4.


Assuntos
Lesão Pulmonar Aguda/imunologia , Exossomos/microbiologia , Ativação de Macrófagos/imunologia , Choque Hemorrágico/imunologia , Receptor 4 Toll-Like/metabolismo , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Animais , Citocinas/biossíntese , Modelos Animais de Doenças , Humanos , Técnicas In Vitro , Mediadores da Inflamação/metabolismo , Linfa/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Choque Hemorrágico/etiologia , Transdução de Sinais , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/deficiência
18.
Am J Respir Cell Mol Biol ; 58(5): 560-565, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29115853

RESUMO

Extracellular vesicles (EVs), such as exosomes and microvesicles, play an important autocrine/paracrine role in intercellular communication. Details on the involvement of EVs in the pathogenesis of lung diseases have emerged over the past several years. Moreover, EVs package numerous DNA, proteins, mRNAs, and microRNAs that can regulate immune responses in recipient cells. Almost all respiratory cells release EVs, and these EVs can have protective or detrimental functions, depending on the type of donor cells, type of stimuli, and components. In lung cancer, tumor-derived EVs carry multiple immunoinhibitory signals, disable antitumor immune effector cells, and promote tumor escape from immune control. Furthermore, bacteria- and microbiota-derived EVs can shape the immune system and lead to the development of lung disease. These EVs are capable of maintaining airway homeostasis, inducing proinflammatory effects, and promoting antigen presentation, thus regulating lung inflammation and immune responses. From these viewpoints, we summarize recent findings on EVs in lung biology and immunity. EVs provide a new avenue for understanding the mechanism of inflammatory disease progression and for developing therapeutic approaches for lung immune responses.


Assuntos
Exossomos/imunologia , Pneumopatias/imunologia , Pulmão/imunologia , Animais , Exossomos/genética , Exossomos/metabolismo , Exossomos/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Pneumopatias/genética , Pneumopatias/metabolismo , Pneumopatias/microbiologia , MicroRNAs/genética , MicroRNAs/imunologia , MicroRNAs/metabolismo , Transdução de Sinais
19.
Adv Exp Med Biol ; 998: 101-112, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28936735

RESUMO

Sepsis is one of the main causes of ICU hospitalization worldwide, with a high mortality rate, and is associated with a large number of comorbidities. One of the main comorbidities associated with sepsis is septic cardiomyopathy. This process occurs mainly due to mechanisms of damage in the cardiovascular system that will lead to changes in cardiovascular physiology, such as decreased Ca2+ response, mitochondrial dysfunction and decreased ß-adrenergic receptor response. Within this process the exosomes play an important role in the pathophysiology of this disease, in which the exosomal content is related to mechanisms that will trigger its development. After platelet activation through ROS exposition, exosomes containing high concentrations of NADPH are released in heart blood vessels, those exosomes will be internalized in endothelial cells leading to cell death and cardiac dysfunction. On the opposite, exosomes derived from mesenchymal stem cells contain miR-223, that have anti-inflammatory properties, are released in less quantities in septic patients causing an imbalance that leads to cardiac dysfunction.


Assuntos
Cardiomiopatias/metabolismo , Exossomos/metabolismo , Miocárdio/metabolismo , Sepse/metabolismo , Transdução de Sinais , Animais , Cardiomiopatias/genética , Cardiomiopatias/microbiologia , Cardiomiopatias/patologia , Exossomos/genética , Exossomos/microbiologia , Exossomos/patologia , Interações Hospedeiro-Patógeno , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Miocárdio/patologia , NADP/metabolismo , Sepse/genética , Sepse/microbiologia , Sepse/patologia
20.
Sci Rep ; 7(1): 6301, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28740179

RESUMO

Extracellular vesicles from eukaryotic cells and outer membrane vesicles (OMVs) released from gram-negative bacteria have been described as mediators of pathogen-host interaction and intercellular communication. Legionella pneumophila (L. pneumophila) is a causative agent of severe pneumonia. The differential effect of bacterial and host cell vesicles in L. pneumophila infection is unknown so far. We infected THP-1-derived or primary human macrophages with L. pneumophila and isolated supernatant vesicles by differential centrifugation. We observed an increase of exosomes in the 100 k pellet by nanoparticle tracking analysis, electron microscopy, and protein markers. This fraction additionally contained Legionella LPS, indicating also the presence of OMVs. In contrast, vesicles in the 16 k pellet, representing microparticles, decreased during infection. The 100 k vesicle fraction activated uninfected primary human alveolar epithelial cells, A549 cells, and THP-1 cells. Epithelial cell activation was reduced by exosome depletion (anti-CD63, or GW4869), or blocking of IL-1ß in the supernatant. In contrast, the response of THP-1 cells to vesicles was reduced by a TLR2-neutralizing antibody, UV-inactivation of bacteria, or - partially - RNase-treatment of vesicles. Taken together, we found that during L. pneumophila infection, neighbouring epithelial cells were predominantly activated by exosomes and cytokines, whereas myeloid cells were activated by bacterial OMVs.


Assuntos
Efeito Espectador , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Interações Hospedeiro-Patógeno , Legionella pneumophila/patogenicidade , Doença dos Legionários/metabolismo , Monócitos/metabolismo , Células A549 , Exossomos/microbiologia , Vesículas Extracelulares/microbiologia , Humanos , Doença dos Legionários/microbiologia , Doença dos Legionários/patologia , Monócitos/microbiologia , Células THP-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA