Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biomedicines ; 11(11)2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-38002055

RESUMEN

The pathogenesis of cerebral small vessel disease (CSVD) is largely unknown. Endothelial disfunction has been suggested as the turning point in CSVD development. In this study, we tested the effect of plasma from CSVD patients on human cerebral microvascular endothelial cells with the aim of describing the pattern of endothelial activation. Plasma samples from three groups of young subjects have been tested: PTs (subjects affected by early stage CSVD); CTRLs (control subjects without abnormalities at MRI scanning); BDs (blood donors). Human Brain Endothelial Cells 5i (HBEC5i) were treated with plasma and total RNA was extracted. RNAs were pooled to reduce gene expression-based variability and NGS analysis was performed. Differentially expressed genes were highlighted comparing PTs, CTRLs and BDs with HBEC5i untreated cells. No significantly altered pathway was evaluated in BD-related treatment. Regulation of p38 MAPK cascade (GO:1900744) was the only pathway altered in CTRL-related treatment. Indeed, 36 different biological processes turned out to be deregulated after PT treatment of HBEC5i, i.e., the cytokine-mediated signaling pathway (GO:0019221). Endothelial cells activate inflammatory pathways in response to stimuli from CSVD patients' plasma, suggesting the pathogenetic role of neuroinflammation from the early asymptomatic phases of cerebrovascular disease.

2.
Sci Rep ; 13(1): 10993, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37419964

RESUMEN

Exosomes are well established effectors of cell-cell communication. Their role on maturation of embryonic cells located in hippocampus, seat of memory, is unknown. Here we show that ceramide facilitates release of exosomes from HN9.10e cells extending information for cell differentiation to neighboring cells. We found only 38 miRNAs differentially expressed in exosomes derived from ceramide-treated cells in comparison with control cells (including 10 up-regulated and 28 down-regulated). Some overexpressed miRNAs (mmu-let-7f-1-3p, mmu-let-7a-1-3p, mmu-let-7b-3p, mmu-let-7b-5p, mmu-miR-330-3p) regulate genes encoding for protein involved in biological, homeostatic, biosynthetic and small molecule metabolic processes, embryo development and cell differentiation, all phenomena relevant for HN9.10e cell differentiation. Notably, the overexpressed mmu-let-7b-5p miRNA appears to be important for our study based on its ability to regulate thirty-five gene targets involved in many processes including sphingolipid metabolism, sphingolipid-related stimulation of cellular functions and neuronal development. Furthermore, we showed that by incubating embryonic cells with exosomes released under ceramide treatment, some cells acquired an astrocytic phenotype and others a neuronal phenotype. We anticipate our study to be a start point for innovative therapeutic strategies to regulate the release of exosomes useful to stimulate delayed brain development in the newborn and to improve the cognitive decline in neurodegenerative disorders.


Asunto(s)
Exosomas , MicroARNs , Exosomas/genética , Exosomas/metabolismo , Ceramidas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Comunicación Celular , Diferenciación Celular/genética
3.
J Clin Med ; 12(3)2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36769731

RESUMEN

OBJECTIVE: To evaluate humoral and T-cell cellular-mediated immune response after three doses of SARS-CoV-2 mRNA vaccines in patients with systemic lupus erythematosus (SLE) under Belimumab. PATIENTS AND METHODS: 12 patients on Belimumab and 13 age-matched healthy volunteers were recruited. Patients were in remission or in low disease activity, and they were taking no corticosteroids or only low doses. None of the patients and controls had detectable anti-SARS-CoV-2 antibodies due to previous exposure to the virus. All the patients received three doses of mRNA anti-SARS-CoV-2 vaccines and the humoral and cellular-mediated response were tested 4 weeks after the second dose (T0), 6 months after the second dose (T1) and 4 weeks after the third dose (T2). Comparison with the control group was performed at time T0 (i.e., 4 weeks after the second dose). Total anti-SARS-CoV-2 RBD antibodies were analyzed using a diagnostic assay, while cellular-mediated response was evaluated using the interferon-gamma release assay (IGRA). RESULTS: A humoral response was documented in all the patients at T0 (median 459; IQR 225.25-758.5), but the antibody titer significantly declined from T0 to T1 (median 44.7; IQR: 30.3-202; p = 0.0066). At T2, the antibody titer significantly increased from T1 (median 2500; IQR: 2500-2500), and it was not different from T0 (respectively p < 0.0001, p = 0.66). Cellular-mediated response significantly declined from T0 to T1 (p = 0.003) but not from T0 to T2 (p = 0.3). No differences were found between patients and controls at T0 as regards both humoral and cellular responses (p = 1.0 and p = 0.09 for humoral and cellular responses, respectively). CONCLUSION: The third dose of mRNA COVID-19 vaccine can restore both humoral and cellular immune response in SLE patients on Belimumab.

6.
J Autoimmun ; 129: 102827, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35427999

RESUMEN

OBJECTIVE: To evaluate B-cell- and T-cell-mediated immune response to SARS-CoV-2 mRNA vaccination in patients with complex or rare systemic autoimmune diseases previously been treated with or under continuous treatment with B-cell-targeted therapies including rituximab (RTX) and belimumab (BEL). MATERIALS AND METHODS: Twenty-eight consecutive patients receiving RTX (n = 11) or BEL (n = 17) treatment and 13 age-/sex-matched controls (non-rheumatic healthcare personnel) were recruited. None of the patients had detectable anti-SARS-CoV-2 antibodies caused by prior exposure to the virus. All the patients and controls received mRNA vaccines and were tested three to four weeks after completion of vaccination. In all the RTX patients, vaccination was started within 5 months from the last infusion, and B-cell depletion was confirmed in all but one of them. Total anti-SARS-CoV-2 RBD antibodies were analyzed using a diagnostic assay, while T-cell response was evaluated using the interferon-gamma release assay (IGRA). Further, SARS-CoV-2 pseudoviruses were employed to verify the strain-specific neutralizing capacity of the antibodies. RESULTS: Detectable anti-SARS-CoV-2 antibodies were documented in 1 out of the 11 RTX patients and 16 of the 17 BEL patients. The median concentration in the RTX and BEL patients was significantly lower than that in the controls (39.6 AU/ml vs. 1133 AU/ml, p = 0.002). The result of IGRA was positive in 8 of the 11 (72.7%) RTX patients and 16 of the 17 (94.1%) BEL patients, and interferon release in both the RTX and BEL patients was comparable to that in the control participants. CONCLUSION: B-cell-targeted therapies do not preclude SARS-CoV-2 vaccination, since virus-specific cellular immunity can be induced even in the absence of circulating B cells. An important finding was that lupus patients treated with BEL developed immune responses to SARS-CoV-2; this indicates retention of the immunogenicity of the COVID-19 vaccine.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Anticuerpos Monoclonales Humanizados , Anticuerpos Antivirales , Humanos , Inmunidad Celular , Rituximab/uso terapéutico , SARS-CoV-2 , Linfocitos T , Vacunación
7.
Biomedicines ; 9(10)2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34680460

RESUMEN

Understanding immune reactivity against SARS-CoV-2 is essential for coping with the COVID-19 pandemic. Herein, we discuss experiences and open questions about the complex immune responses to SARS-CoV-2. Some people react excellently without experiencing any clinical symptoms, they do not get sick, and they do not pass the virus on to anyone else ("sterilizing" immunity). Others produce antibodies and do not get COVID-19 but transmit the virus to others ("protective" immunity). Some people get sick but recover. A varying percentage develops respiratory failure, systemic symptoms, clotting disorders, cytokine storms, or multi-organ failure; they subsequently decease. Some develop long COVID, a new pathologic entity similar to fatigue syndrome or autoimmunity. In reality, COVID-19 is considered more of a systemic immune-vascular disease than a pulmonic disease, involving many tissues and the central nervous system. To fully comprehend the complex clinical manifestations, a profound understanding of the immune responses to SARS-CoV-2 is a good way to improve clinical management of COVID-19. Although neutralizing antibodies are an established approach to recognize an immune status, cellular immunity plays at least an equivalent or an even more important role. However, reliable methods to estimate the SARS-CoV-2-specific T cell capacity are not available for clinical routines. This deficit is important because an unknown percentage of people may exist with good memory T cell responsibility but a low number of or completely lacking peripheral antibodies against SARS-CoV-2. Apart from natural immune responses, vaccination against SARS-CoV-2 turned out to be very effective and much safer than naturally acquired immunity. Nevertheless, besides unwanted side effects of the currently available vector and mRNA preparations, concerns remain whether these vaccines will be strong enough to defeat the pandemic. Altogether, herein we discuss important questions, and try to give answers based on the current knowledge and preliminary data from our laboratories.

8.
Oncol Rep ; 46(3)2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34318905

RESUMEN

Anaplastic thyroid cancer (ATC) is the most aggressive type of thyroid cancer and is responsible for 20­50% of thyroid cancer­associated deaths. The absence of response to conventional treatments makes the search for novel therapeutics a clinical challenge. In the present study, the effects of 15,16­dihydrotanshinone I (DHT), a tanshinone extracted from Salvia miltiorrhiza Bunge (Danshen), which has previously been shown to possess anticancer activity, were examined in two human ATC cell lines. DHT significantly reduced cell viability, which was coupled with an increase in apoptosis. DHT administration also reduced the colony­forming ability and proliferation of these cells in soft agar and downregulated the expression of epithelial­to­mesenchymal transition­related genes. In addition, DHT significantly reduced MAD2 expression, a target of HuR with a relevant role in ATC. Finally, cotreatment with cisplatin and DHT has a greater effect on cell viability than each compound alone. In conclusion, to the best of our knowledge, the present study is the first to demonstrate that DHT exerts antitumor effects on ATC cells by reducing MAD2 expression levels. Moreover, a synergistic effect of DHT with cisplatin was shown. Further in vivo studies are required to assess this phytochemical compound as a potential adjuvant for the treatment of ATC.


Asunto(s)
Cisplatino/farmacología , Furanos/farmacología , Fenantrenos/farmacología , Quinonas/farmacología , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos
9.
Int J Mol Sci ; 22(12)2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-34200790

RESUMEN

Exosomes released from tumor cells are instrumental in shaping the local tumor microenvironment to allow cancer progression. Recently, it has been shown that tumor exosomes carry large fragments of dsDNA, which may reflect the mutational status of parental cells. Although it has been described that a stressful microenvironment can influence exosomal cargo, the effects on DNA packing and its transfer into recipient cells have yet to be investigated. Here, we report that exosomes derived from SW480 (human colorectal adenocarcinoma cell line) cells can carry dsDNA fragments containing the entire coding sequence of both TP53 and KRAS genes, harboring the SW480-related TP53 c.818G > A and KRAS c.35G > T typical mutations. We also report the following: that cell stimulation with lipopolysaccharides (LPS) promotes the selective packaging of the TP53 gene, but not the KRAS gene; that exosomes secreted by SW480 cells efficiently transfer the mutated sequences into normal CCD841-CoN colon epithelial and THLE-2 hepatic cells; that this mechanism is more efficient when the cells had been previously incubated with pro-inflammatory cytokines; that the TP53 gene appears actively transcribed in both recipient cells; and that mutated mRNA levels are not influenced by cytokine treatment. Our data strongly suggest that pro-inflammatory stimulation promotes the horizontal transfer of an oncogene by exosomes, although this remains a rare event. Further studies are needed to assess the impact of the oncogenic transfer by exosomes in malignant transformation and its role in tumor progression.


Asunto(s)
Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/patología , Exosomas/genética , Mediadores de Inflamación/inmunología , Mutación , Microambiente Tumoral/inmunología , Proteína p53 Supresora de Tumor/genética , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/genética , Humanos , Células Tumorales Cultivadas
10.
Cytokine ; 140: 155438, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33493861

RESUMEN

BACKGROUND: Patients infected by SARS-CoV-2 can develop interstitial pneumonia, requiring hospitalisation or mechanical ventilation. Increased levels of inflammatory biomarkers are associated with development of acute respiratory distress syndrome (ARDS). The aim of the present study was to determine which cytokines are associated with respiratory insufficiency in patients hospitalised for COVID-19. PATIENTS AND METHODS: Data on 67 consecutive patients were collected between March 8 and March 30, 2020. PaO2/FiO2 ratio (P/F) was calculated at hospital admission. The following cytokines were analysed: interleukin (IL)-6, IL-1α, IL-18, tumour necrosis factor (TNF)-ß, macrophage colony-stimulating factor (M-CSF), macrophage migration inhibitory factor (MIF), soluble IL-2 receptor alpha (sIL-2Rα; CD25), IL-12ß, IL-3, interferon (IFN) α2a, monokine induced by gamma interferon (MIG), monocyte-chemotactic protein 3 (MCP3) and hepatocyte growth factor (HGF). RESULTS: P/F lower than 300 was recorded in 22 out of 67 patients (32.8%). P/F strongly correlated with IL-6 (r = -0.62, P < 0.0001), M-CSF (r = -0.63, P < 0.0001), sIL-2Rα (r = -0.54, P < 0.0001), and HGF (r = -0.53, P < 0.0001). ROC curve analyses for IL-6 (AUC 0.83, 95% CI 0.73-0.93, P < 0.0001), M-CSF (AUC 0.87, 95% CI 0.79-0.96, P < 0.0001), HGF (AUC 0.81, 95% CI 0.70-0.93, P < 0.0001), and sIL-2Rα (AUC 0.80, 95% CI, 0.69-0.90, P < 0.0001) showed that these four soluble factors were highly significant. All four soluble factors correlated with LDH, white blood cell count, neutrophil count, lymphocyte count, and CRP. CONCLUSION: IL-6, M-CSF, sIL-2Rα, and HGF are possibly involved in the main biological processes of severe COVID-19, mirroring the level of systemic hyperinflammatory state, the level of lung inflammation, and the severity of organ damage.


Asunto(s)
COVID-19/sangre , Citocinas/sangre , Inmunidad Innata/inmunología , Inflamación/sangre , Subunidad alfa del Receptor de Interleucina-2/sangre , Insuficiencia Multiorgánica/sangre , Neumonía/sangre , Anciano , COVID-19/complicaciones , COVID-19/virología , Femenino , Factor de Crecimiento de Hepatocito/sangre , Interacciones Huésped-Patógeno , Humanos , Inflamación/complicaciones , Interleucina-6/sangre , Factor Estimulante de Colonias de Macrófagos/sangre , Masculino , Persona de Mediana Edad , Insuficiencia Multiorgánica/complicaciones , Neumonía/complicaciones , Neumonía/virología , Estudios Retrospectivos , SARS-CoV-2/fisiología
11.
Int J Mol Sci ; 21(22)2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33227947

RESUMEN

Exosomes secreted by tumor cells, through the transport of bioactive molecules, reprogram the surroundings, building a microenvironment to support the development of the tumor. The discovery that exosomes carry genomic DNA reflecting that of the tumor cell of origin has encouraged studies to use them as non-invasive biomarkers. The exosome-mediated transfer of oncogenes suggested a new mechanism of malignant transformation that could play a role in the formation of metastases. Several studies have examined the role of tumor exosomes on the modulation of the tumor microenvironment, but relatively few have been directed to assess how stressful stimuli can influence their production and cargo. Understanding the changes in exosome loads and the production pattern of the stressed tumor cell may uncover actionable mechanisms responsible for tumor progression.


Asunto(s)
Biomarcadores de Tumor/genética , ADN Tumoral Circulante/genética , Exosomas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Neoplasias/genética , Transporte Biológico , Biomarcadores de Tumor/sangre , Comunicación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , ADN Tumoral Circulante/sangre , Exosomas/química , Exosomas/patología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Metástasis Linfática , Proteínas de Neoplasias/metabolismo , Neoplasias/sangre , Neoplasias/patología , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Oncogenes , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Estrés Fisiológico , Microambiente Tumoral/genética
12.
PLoS One ; 15(11): e0242342, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33180848

RESUMEN

Vedolizumab (VDZ) is a therapeutic monoclonal antibody approved for the treatment of inflammatory bowel diseases (IBD). VDZ selectively binds to the α4ß7 integrin and blocks trafficking of a specific subset of gastrointestinal-homing T-lymphocytes to inflamed tissue. Although VDZ has shown promising results in numerous clinical studies a subgroup of patients do not respond adequately. Mechanistic insights and prognostic biomarkers able to predict which patients might benefit from VDZ therapy are currently lacking. Circulating exosomes were isolated from serum of blood donors and VDZ-treated patients by polymer-based precipitation. The surface expression of α4ß7 integrin was evaluated by flow cytometry and the levels of exosome-bound VDZ were investigated by Promonitor-VDZ ELISA kit. The capacity of exosomes to interfere with the adhesion of VDZ-treated CD4+ T cells was assessed by adhesion assay. In this study, we showed that serum exosomes isolated from both blood donor and ulcerative colitis patients express on their surface the VDZ target α4ß7 integrin. We observed an increased exosomal sequestration of VDZ in anti-TNF exposed patients compared to anti- TNFα naïve patients, according to a greater expression of α4ß7 integrin on vesicles surface. Circulating exosomes could compete for VDZ binding with CD4+ T cells since we found that the amount of VDZ bound to T cells was impaired in the presence of exosomes. In addition, we demonstrated that exosomes bind VDZ, which consequently becomes unable to block MadCAM-1-mediated adhesion of lymphocytes. Circulating exosomes might contribute to drug sequestration, possibly affecting the therapeutic efficacy of VDZ in IBD patients. Our data suggest that previous biologic therapy may have altered the sequestration capacity of circulating exosomes, thus reducing the efficacy of VDZ in patients who failed anti-TNF agents.


Asunto(s)
Anticuerpos Monoclonales Humanizados/metabolismo , Exosomas/genética , Integrinas/genética , Adulto , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Biomarcadores Farmacológicos/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Colitis Ulcerosa/tratamiento farmacológico , Exosomas/metabolismo , Femenino , Fármacos Gastrointestinales/uso terapéutico , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Integrinas/metabolismo , Masculino , Persona de Mediana Edad , Unión Proteica/genética , Unión Proteica/fisiología , Estudios Retrospectivos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/uso terapéutico
13.
Stem Cells Int ; 2020: 4058760, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733572

RESUMEN

OBJECTIVE: Several clinical studies have proposed the infusion of adipose mesenchymal stem cells (AMSCs) as an alternative therapy for joint diseases with inflammatory components, such as osteoarthritis. Indeed, AMSCs are able to stimulate tissue repair through a paracrine activity and the interaction with the inflammatory microenvironment seems to have a critical role. DESIGN: To reproduce the inflammatory microenvironment, AMSCs were exposed to osteoarthritic synovial fluid (SF) for 48 h and the effect of their secretome on differentiation of monocytes (M0) into macrophages M1-like and mature dendritic cells (mDCs) was evaluated. Furthermore, the effect of the secretome of AMSCs exposed to SF was evaluated on the T cell population in terms of T cell proliferation and expansion of T regulatory cells (T reg). RESULTS: Our data show that the exposure of AMSCs to SF activates cells and promotes the release of immunosuppressive factors, which induce macrophage polarization of M0 into the M2-like phenotype and inhibit differentiation of monocytes into mature dendritic cells (mDCs). Only the secretome of exposed AMSCs was able to inhibit T cell proliferation and promote T reg expansion. CONCLUSIONS: Our results suggest that the microenvironment plays a fundamental role for the development of anti-inflammatory and immunomodulatory properties of AMSCs.

14.
Sci Rep ; 9(1): 14207, 2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31578412

RESUMEN

Sporadic small vessel disease (SVD) has high prevalence in aging population and stroke patients, but also in younger asymptomatic subjects. In this last group it can represents a prelude to stroke and cognitive impairment. Still nowadays, its pathogenesis is unclear. 35 consecutive patients with SVD at brain MRI and 35 age- and sex-matched controls, between January 2016 and February 2018, underwent an extended screening for thrombophilia, autoimmunity and evaluated levels of blood markers of inflammation and endothelial activation. Asymmetric DiMethyl Arginine (ADMA) levels proved higher in patients (70.44 ± 36.25 ng/ml vs. 46.58 ± 30.67 ng/ml; p = 0.004), also after controlling for confounding factors. ADMA levels showed positive correlation with Fazekas score (r = 0.304; p = 0.01). ROC curve analysis showed a moderate accuracy in discriminating patients and controls (AUC = 0.70; CI 0.57-0.82; p = 0.004): a cut-off of 46 ng/ml is associated with 80% sensitivity, but limited (54%) specificity. Higher ADMA levels characterize selected subjects with sporadic SVD, asymptomatic for vascular diseases and without latent inflammatory conditions or coagulopathy. This reinforces the hypothesis of the key role of endothelial dysfunction in SVD. Further studies should explore the cause-effect relationship between ADMA pathway and SVD.


Asunto(s)
Arginina/sangre , Biomarcadores/sangre , Enfermedades de los Pequeños Vasos Cerebrales/sangre , Accidente Cerebrovascular/sangre , Adulto , Anciano , Envejecimiento/sangre , Envejecimiento/patología , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Enfermedades de los Pequeños Vasos Cerebrales/diagnóstico por imagen , Enfermedades de los Pequeños Vasos Cerebrales/patología , Disfunción Cognitiva/sangre , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/patología
15.
Sci Rep ; 9(1): 8457, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186484

RESUMEN

The biology of tumor-derived exosomes (TEX) is only partially understood and much remains to be studied in order to define the effect that the tumor microenvironment or the activation of tumor cells exerts on their composition and functions. Increased expression and activity of toll-like receptor 4 (TLR4) in chronic infectious and inflammatory conditions is related with cancer progression: its activation induces an inflammatory signaling that increases the tumorigenic potential of cancer cells promoting their immune evasion. We investigated the immune modulatory properties of TEX released upon cell TLR4 activation, and we found that, although differences were observed depending on the type of the tumor, the treatment influences TEX composition and boosts their immunosuppressive ability. Our results suggest that the activation of TLR4 supports tumor progression by stimulating the release of more effective immunosuppressive exosomes, which allow tumor cells to escape immune surveillance and probably even play a role in the metastatic process.


Asunto(s)
Exosomas/genética , Neoplasias/genética , Receptor Toll-Like 4/genética , Microambiente Tumoral/inmunología , Línea Celular Tumoral , Exosomas/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunosupresores/inmunología , Inmunosupresores/metabolismo , MicroARNs/genética , Neoplasias/clasificación , Neoplasias/inmunología , Neoplasias/patología , Transducción de Señal , Receptor Toll-Like 4/inmunología , Microambiente Tumoral/genética
16.
Sci Rep ; 8(1): 13325, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30190615

RESUMEN

The predominant mechanism by which adipose mesenchymal stem cells (AMSCs) participate to tissue repair is through a paracrine activity and their communication with the inflammatory microenvironment is essential part of this process. This hypothesis has been strengthened by the recent discovery that stem cells release not only soluble factors but also extracellular vesicles, which elicit similar biological activity to the stem cells themselves. We demonstrated that the treatment with inflammatory cytokines increases the immunosuppressive and anti-inflammatory potential of AMSCs-derived exosomes, which acquire the ability to shift macrophages from M1 to M2 phenotype by shuttling miRNA regulating macrophages polarization. This suggests that the immunomodulatory properties of AMSCs-derived exosomes may be not constitutive, but are instead induced by the inflammatory microenvironment.


Asunto(s)
Tejido Adiposo/inmunología , Microambiente Celular/inmunología , Exosomas/inmunología , Tolerancia Inmunológica , Macrófagos/inmunología , Células Madre Mesenquimatosas/inmunología , Humanos , Inflamación/inmunología
17.
Cell Immunol ; 332: 85-93, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30097176

RESUMEN

S100A4 protein is expressed in fibroblasts during tissue remodelling and in cancer stem cells and it induces the metastatic spread of tumor cells. In mast cells (MCs) S100A4 have been found in some pathological conditions, but its function in normal MCs remains to be described. The purpose of this study was to characterize the cellular localization of the S100A4 protein in MCs of human tissues with inflammatory or tumor disorders and, to determine the consequence of reducing its expression in MC response. We found that tissue resident MCs stained positive to S100A4. Both human HMC-1 cell line and resting CD34+-derived MCs expressed S100A4, whose levels were differentially modulated upon MC activation. Downregulation of the S100A4 protein resulted in MC growth inhibition, enhanced apoptosis and deregulation of MMP-1 and MMP-10 production. Our results suggest that S100A4 is also playing a role in the MC life cycle and functions.


Asunto(s)
Mastocitos/metabolismo , Proteína de Unión al Calcio S100A4/metabolismo , Antígenos CD34/metabolismo , Apoptosis/fisiología , Células Cultivadas , Regulación hacia Abajo/fisiología , Fibroblastos/metabolismo , Humanos , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 10 de la Matriz/metabolismo , Células Madre Neoplásicas/metabolismo
18.
Mediators Inflamm ; 2017: 4814987, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28634420

RESUMEN

The purpose of this study is to characterize synovial fluid- (SF-) derived exosomes of patients with gonarthrosis comparing two methods of isolation and to investigate their immune regulatory properties. Extracellular vesicles (EVs) have been isolated from inflamed SF by polymer precipitation method and quantified by Exocet kit and by nanoparticle tracking analysis. Vesicles expressed all the specific exosomal markers by immunoblot and FACS. After isolation with Exoquick, a relevant contamination by immune complexes was detected, which required further magnetic bead-based purification to remove. SF-derived exosomes significantly stimulated the release of several inflammatory cytokines and chemokines and metalloproteinases by M1 macrophages but did not influence the expression of CD80 and CD86 costimulatory molecules. In conclusion, we characterized purified exosomes isolated from inflamed SF and demonstrate that purified exosomes are functionally active in their ability to stimulate the release of proinflammatory factors from M1 macrophages. Our data indicate that SF-derived exosomes from gonarthrosis patients play a role in disease progression.


Asunto(s)
Exosomas/metabolismo , Osteoartritis/metabolismo , Líquido Sinovial/metabolismo , Anciano , Anciano de 80 o más Años , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Exosomas/química , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad
19.
PLoS One ; 12(1): e0169932, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28107450

RESUMEN

A major contributing factor to glioma development and progression is its ability to evade the immune system. Nano-meter sized vesicles, exosomes, secreted by glioma-stem cells (GSC) can act as mediators of intercellular communication to promote tumor immune escape. Here, we investigated the immunomodulatory properties of GCS-derived exosomes on different peripheral immune cell populations. Healthy donor peripheral blood mononuclear cells (PBMCs) stimulated with anti-CD3, anti-CD28 and IL-2, were treated with GSC-derived exosomes. Phenotypic characterization, cell proliferation, Th1/Th2 cytokine secretion and intracellular cytokine production were analysed by distinguishing among effector T cells, regulatory T cells and monocytes. In unfractionated PBMCs, GSC-derived exosomes inhibited T cell activation (CD25 and CD69 expression), proliferation and Th1 cytokine production, and did not affect cell viability or regulatory T-cell suppression ability. Furthermore, exosomes were able to enhance proliferation of purified CD4+ T cells. In PBMCs culture, glioma-derived exosomes directly promoted IL-10 and arginase-1 production and downregulation of HLA-DR by unstimulated CD14+ monocytic cells, that displayed an immunophenotype resembling that of monocytic myeloid-derived suppressor cells (Mo-MDSCs). Importantly, the removal of CD14+ monocytic cell fraction from PBMCs restored T-cell proliferation. The same results were observed with exosomes purified from plasma of glioblastoma patients. Our results indicate that glioma-derived exosomes suppress T-cell immune response by acting on monocyte maturation rather than on direct interaction with T cells. Selective targeting of Mo-MDSC to treat glioma should be considered with regard to how immune cells allow the acquirement of effector functions and therefore counteracting tumor progression.


Asunto(s)
Exosomas/inmunología , Terapia de Inmunosupresión , Monocitos/inmunología , Células Madre Neoplásicas/inmunología , Linfocitos T Reguladores/inmunología , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Citometría de Flujo , Glioblastoma/sangre , Glioblastoma/inmunología , Humanos , Activación de Linfocitos
20.
Lab Invest ; 96(9): 959-971, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27348627

RESUMEN

Endometriosis is an inflammatory disease characterized by the presence of ectopic endometrial tissue outside the uterus. A diffuse infiltration of mast cells (MCs) is observed throughout endometriotic lesions, but little is known about how these cells contribute to the network of molecules that modulate the growth of ectopic endometrial implants and promote endometriosis-associated inflammation. The aryl hydrocarbon receptor (AhR), a transcription factor known to respond to environmental toxins and endogenous compounds, is present in MCs. In response to AhR activation, MCs produce IL-17 and reactive oxygen species, highlighting the potential impact of AhR ligands on inflammation via MCs. Here, we investigated the possibility that endometrial MCs promote an inflammatory microenvironment by sensing AhR ligands, thus sustaining endometriosis development. Using human endometriotic tissue (ET) samples, we performed the following experiments: (i) examined the cytokine expression profile; (ii) counted AhR-expressing MCs; (iii) verified the phenotype of AhR-expressing MCs to establish whether MCs have a tolerogenic (IL-10-positive) or inflammatory (IL-17-positive) phenotype; (iv) measured the presence of AhR ligands (tryptophan-derived kynurenine) and tryptophan-metabolizing enzymes (indoleamine 2,3-dioxygenase 1 (IDO1)); (v) treated ET organ cultures with an AhR antagonist in vitro to measure changes in the cytokine milieu; and (vi) measured the growth of endometrial stromal cells cultured with AhR-activated MC-conditioned medium. We found that ET tissue was conducive to cytokine production, orchestrating chronic inflammation and a population of AhR-expressing MCs that are both IL-17 and IL-10-positive. ET was rich in IDO1 and the AhR-ligand kynurenine compared with control tissue, possibly promoting MC activation through AhR. ET was susceptible to treatment with an AhR antagonist, and endometrial stromal cell growth was improved in the presence of soluble factors released by MCs on AhR activation. These results suggest a new mechanistic role of MCs in the pathogenesis of endometriosis.


Asunto(s)
Citocinas/metabolismo , Endometriosis/metabolismo , Mastocitos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Células Cultivadas , Femenino , Humanos , Inmunohistoquímica , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Quinurenina/metabolismo , Ligandos , Microscopía Fluorescente , Persona de Mediana Edad , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA