Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 17(4): e1009505, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33857267

RESUMEN

The Tick-borne encephalitis virus (TBEV) causes different disease symptoms varying from asymptomatic infection to severe encephalitis and meningitis suggesting a crucial role of the human host immune system in determining the fate of the infection. There is a need to understand the mechanisms underpinning TBEV-host interactions leading to protective immunity. To this aim, we studied the response of human peripheral blood mononuclear cells (PBMC) to the whole formaldehyde inactivated TBEV (I-TBEV), the drug substance of Encepur, one of the five commercially available vaccine. Immunophenotyping, transcriptome and cytokine profiling of PBMC revealed that I-TBEV generates differentiation of a sub-population of plasmacytoid dendritic cells (pDC) that is specialized in type I interferon (IFN) production. In contrast, likely due to the presence of aluminum hydroxide, Encepur vaccine was a poor pDC stimulus. We demonstrated I-TBEV-induced type I IFN together with Interleukin 6 and BAFF to be critical for B cell differentiation to plasmablasts as measured by immunophenotyping and immunoglobulin production. Robust type I IFN secretion was induced by pDC with the concerted action of both viral E glycoprotein and RNA mirroring previous data on dual stimulation of pDC by both S. aureus and influenza virus protein and nucleic acid that leads to a type I IFN-mediated sustained immune response. E glycoprotein neutralization or high temperature denaturation and inhibition of Toll-like receptor 7 signalling confirmed the importance of preserving the functional integrity of these key viral molecules during the inactivation procedure and manufacturing process to produce a vaccine able to stimulate strong immune responses.


Asunto(s)
Células Dendríticas/inmunología , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/prevención & control , Interacciones Microbiota-Huesped , Interferón Tipo I/metabolismo , Vacunas Virales/inmunología , Antivirales/inmunología , Diferenciación Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Dendríticas/virología , Encefalitis Transmitida por Garrapatas/virología , Humanos , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , ARN Viral/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
2.
PLoS Pathog ; 17(9): e1009878, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34473805

RESUMEN

SARS-CoV-2 fine-tunes the interferon (IFN)-induced antiviral responses, which play a key role in preventing coronavirus disease 2019 (COVID-19) progression. Indeed, critically ill patients show an impaired type I IFN response accompanied by elevated inflammatory cytokine and chemokine levels, responsible for cell and tissue damage and associated multi-organ failure. Here, the early interaction between SARS-CoV-2 and immune cells was investigated by interrogating an in vitro human peripheral blood mononuclear cell (PBMC)-based experimental model. We found that, even in absence of a productive viral replication, the virus mediates a vigorous TLR7/8-dependent production of both type I and III IFNs and inflammatory cytokines and chemokines, known to contribute to the cytokine storm observed in COVID-19. Interestingly, we observed how virus-induced type I IFN secreted by PBMC enhances anti-viral response in infected lung epithelial cells, thus, inhibiting viral replication. This type I IFN was released by plasmacytoid dendritic cells (pDC) via an ACE-2-indipendent but Neuropilin-1-dependent mechanism. Viral sensing regulates pDC phenotype by inducing cell surface expression of PD-L1 marker, a feature of type I IFN producing cells. Coherently to what observed in vitro, asymptomatic SARS-CoV-2 infected subjects displayed a similar pDC phenotype associated to a very high serum type I IFN level and induction of anti-viral IFN-stimulated genes in PBMC. Conversely, hospitalized patients with severe COVID-19 display very low frequency of circulating pDC with an inflammatory phenotype and high levels of chemokines and pro-inflammatory cytokines in serum. This study further shed light on the early events resulting from the interaction between SARS-CoV-2 and immune cells occurring in vitro and confirmed ex vivo. These observations can improve our understanding on the contribution of pDC/type I IFN axis in the regulation of the anti-viral state in asymptomatic and severe COVID-19 patients.


Asunto(s)
COVID-19/inmunología , Células Dendríticas/clasificación , Interferón Tipo I/metabolismo , SARS-CoV-2/inmunología , Adulto , Anciano de 80 o más Años , Infecciones Asintomáticas , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/virología , Células Epiteliales/citología , Femenino , Hospitalización , Humanos , Interferón Tipo I/inmunología , Pulmón/citología , Masculino , Persona de Mediana Edad , Neuropilina-1/metabolismo , Fenotipo , Índice de Severidad de la Enfermedad , Receptor Toll-Like 7/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(28): E6546-E6555, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29941600

RESUMEN

The tumor microenvironment restrains conventional T cell (Tconv) activation while facilitating the expansion of Tregs. Here we showed that Tregs' advantage in the tumor milieu relies on supplemental energetic routes involving lipid metabolism. In murine models, tumor-infiltrating Tregs displayed intracellular lipid accumulation, which was attributable to an increased rate of fatty acid (FA) synthesis. Since the relative advantage in glucose uptake may fuel FA synthesis in intratumoral Tregs, we demonstrated that both glycolytic and oxidative metabolism contribute to Tregs' expansion. We corroborated our data in human tumors showing that Tregs displayed a gene signature oriented toward glycolysis and lipid synthesis. Our data support a model in which signals from the tumor microenvironment induce a circuitry of glycolysis, FA synthesis, and oxidation that confers a preferential proliferative advantage to Tregs, whose targeting might represent a strategy for cancer treatment.


Asunto(s)
Ácidos Grasos/inmunología , Glucólisis/inmunología , Neoplasias Experimentales/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/inmunología , Ácidos Grasos/genética , Humanos , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Oxidación-Reducción , Linfocitos T Reguladores/patología , Microambiente Tumoral/genética
4.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209845

RESUMEN

The vulnerability of humankind to SARS-CoV-2 in the absence of a pre-existing immunity, the unpredictability of the infection outcome, and the high transmissibility, broad tissue tropism, and ability to exploit and subvert the immune response pose a major challenge and are likely perpetuating the COVID-19 pandemic. Nevertheless, this peculiar infectious scenario provides researchers with a unique opportunity for studying, with the latest immunological techniques and understandings, the immune response in SARS-CoV-2 naïve versus recovered subjects as well as in SARS-CoV-2 vaccinees. Interestingly, the current understanding of COVID-19 indicates that the combined action of innate immune cells, cytokines, and chemokines fine-tunes the outcome of SARS-CoV-2 infection and the related immunopathogenesis. Indeed, the emerging picture clearly shows that the excessive inflammatory response against this virus is among the main causes of disease severity in COVID-19 patients. In this review, the innate immune response to SARS-CoV-2 infection is described not only in light of its capacity to influence the adaptive immune response towards a protective phenotype but also with the intent to point out the multiple strategies exploited by SARS-CoV-2 to antagonize host antiviral response and, finally, to outline inborn errors predisposing individuals to COVID-19 disease severity.


Asunto(s)
COVID-19/patología , Inmunidad Innata , COVID-19/inmunología , COVID-19/virología , Quimiocinas/metabolismo , Citocinas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Monocitos/citología , Monocitos/metabolismo , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad
5.
PLoS Pathog ; 14(1): e1006790, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29300789

RESUMEN

Autophagy is a primordial eukaryotic pathway, which provides the immune system with multiple mechanisms for the elimination of invading pathogens including Mycobacterium tuberculosis (Mtb). As a consequence, Mtb has evolved different strategies to hijack the autophagy process. Given the crucial role of human primary dendritic cells (DC) in host immunity control, we characterized Mtb-DC interplay by studying the contribution of cellular microRNAs (miRNAs) in the post-transcriptional regulation of autophagy related genes. From the expression profile of de-regulated miRNAs obtained in Mtb-infected human DC, we identified 7 miRNAs whose expression was previously found to be altered in specimens of TB patients. Among them, gene ontology analysis showed that miR-155, miR-155* and miR-146a target mRNAs with a significant enrichment in biological processes linked to autophagy. Interestingly, miR-155 was significantly stimulated by live and virulent Mtb and enriched in polysome-associated RNA fraction, where actively translated mRNAs reside. The putative pair interaction among the E2 conjugating enzyme involved in LC3-lipidation and autophagosome formation-ATG3-and miR-155 arose by target prediction analysis, was confirmed by both luciferase reporter assay and Atg3 immunoblotting analysis of miR-155-transfected DC, which showed also a consistent Atg3 protein and LC3 lipidated form reduction. Late in infection, when miR-155 expression peaked, both the level of Atg3 and the number of LC3 puncta per cell (autophagosomes) decreased dramatically. In accordance, miR-155 silencing rescued autophagosome number in Mtb infected DC and enhanced autolysosome fusion, thereby supporting a previously unidentified role of the miR-155 as inhibitor of ATG3 expression. Taken together, our findings suggest how Mtb can manipulate cellular miRNA expression to regulate Atg3 for its own survival, and highlight the importance to develop novel therapeutic strategies against tuberculosis that would boost autophagy.


Asunto(s)
Proteínas Relacionadas con la Autofagia/genética , Autofagia/genética , Células Dendríticas/metabolismo , MicroARNs/genética , Mycobacterium tuberculosis/fisiología , Enzimas Ubiquitina-Conjugadoras/genética , Autofagosomas/inmunología , Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/antagonistas & inhibidores , Células Cultivadas , Células Dendríticas/microbiología , Regulación de la Expresión Génica , Células HEK293 , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , MicroARNs/fisiología , Mycobacterium tuberculosis/inmunología , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores
6.
J Autoimmun ; 101: 1-16, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31047767

RESUMEN

Alteration in endogenous Interferon (IFN) system may profoundly impact immune cell function in autoimmune diseases. Here, we provide evidence that dysregulation in IFN-regulated genes and pathways are involved in B cell- and monocyte-driven pathogenic contribution to Multiple Sclerosis (MS) development and maintenance. In particular, by using an Interferome-based cell type-specific approach, we characterized an increased susceptibility to an IFN-linked caspase-3 dependent apoptotic cell death in both B cells and monocytes of MS patients that may arise from their chronic activation and persistent stimulation by activated T cells. Ongoing caspase-3 activation functionally impacts on MS monocyte properties influencing the STAT-3/IL-16 axis, thus, driving increased expression and massive release of the bio-active IL-16 triggering and perpetuating CD4+ T cell migration. Importantly, our analysis also identified a previously unknown multi-component defect in type I IFN-mediated signaling and response to virus pathways specific of MS B cells, impacting on induction of anti-viral responses and Epstein-barr virus infection control in patients. Taking advantage of cell type-specific transcriptomics and in-depth functional validation, this study revealed pathogenic contribution of endogenous IFN signaling and IFN-regulated cell processes to MS pathogenesis with implications on fate and functions of B cells and monocytes that may hold therapeutic potential.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Interferón Tipo I/genética , Monocitos/inmunología , Monocitos/metabolismo , Esclerosis Múltiple/etiología , Esclerosis Múltiple/metabolismo , Transcriptoma , Adulto , Apoptosis , Biomarcadores , Estudios de Casos y Controles , Susceptibilidad a Enfermedades , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Interferón Tipo I/metabolismo , Interleucina-16/genética , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/patología , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Regiones Promotoras Genéticas , Transducción de Señal
7.
Mult Scler ; 24(2): 127-139, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28273784

RESUMEN

BACKGROUND: B cells are key pathogenic effectors in multiple sclerosis (MS) and several therapies have been designed to restrain B cell abnormalities by directly targeting this lymphocyte population. OBJECTIVES: Moving from our data showing a Toll-like receptor (TLR)7-driven dysregulation of B cell response in relapsing-remitting multiple sclerosis (RRMS) and having found a low serum level of Thymosin-α1 (Tα1) in patients, we investigated whether the addition of this molecule to peripheral blood mononuclear cells (PBMCs) would influence the expansion of regulatory B cell subsets, known to dampen autoimmune inflammation. METHODS: Serum Tα1 level was measured by enzyme immunoassay. Cytokine expression was evaluated by Cytometric Bead Array (CBA), enzyme-linked immunosorbent assay (ELISA), and real-time reverse transcription polymerase chain reaction (RT-PCR). B cell subsets were analyzed by flow cytometry. RESULTS: Tα1 pre-treatment induces an anti-inflammatory status in TLR7-stimulated RRMS PBMC cultures, reducing the secretion of pro-inflammatory interleukin (IL)-6, IL-8, and IL-1ß while significantly increasing the regulatory IL-10 and IL-35. Indeed, Tα1 treatment enhanced expansion of CD19+CD24+CD38hi transitional-immature and CD24low/negCD38hi plasmablast-like regulatory B cell subsets, which likely inhibit both interferon (IFN)-γ and IL-17 production. CONCLUSION: Our study reveals a deficient ability of B cells from MS patients to differentiate into regulatory subsets and unveils a novel anti-inflammatory and repurposing potential for Tα1 in MS targeting B cell response.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Linfocitos B Reguladores/efectos de los fármacos , Citocinas/metabolismo , Interleucina-10/metabolismo , Esclerosis Múltiple Recurrente-Remitente/sangre , Timalfasina/sangre , Timalfasina/farmacología , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptor Toll-Like 7/agonistas , Adulto Joven
8.
Semin Immunol ; 26(6): 543-51, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25453229

RESUMEN

A major challenge in tuberculosis (TB) is to improve current vaccination and therapeutic strategies and this requires a fine understanding of the mechanisms that mediate protection and pathogenesis. We need to discern how the host perceives Mycobacterium tuberculosis (Mtb) infection, what are the danger signals that activate the immune system and, in turn, how the immune response controls the life-cycle of Mtb. Cytokines, because of their nature of soluble mediators, represent key elements in mediating and tuning these complex processes. In this review, we provide an overview of recent studies on cytokines expression and function in active (mainly human) TB. Understanding of the balance between pro- and anti-inflammatory networks is crucial to refine our knowledge on the immune responses directed against Mtb.


Asunto(s)
Citocinas/inmunología , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/microbiología , Células Epiteliales Alveolares/patología , Citocinas/genética , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Células Dendríticas/patología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Pulmón/microbiología , Pulmón/patología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/microbiología , Linfocitos T/patología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
9.
J Infect Dis ; 213(4): 674-83, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26386427

RESUMEN

CD8(+) T cells specific to caspase-cleaved antigens derived from apoptotic T cells represent a principal player in chronic immune activation. Here, we found that both apoptotic epitope-specific and hepatitis C virus (HCV)-specific CD8(+) T cells were mostly confined within the effector memory (EM) or terminally differentiated EM CD45RA(+) cell subsets expressing a dysfunctional T-helper 1-like signature program in chronic HCV infection. However, apoptotic epitope-specific CD8(+) T cells produced tumor necrosis factor α and interleukin 2 at the intrahepatic level significantly more than HCV-specific CD8(+) T cells, despite both populations expressing high levels of programmed death 1 receptor. Contextually, only apoptotic epitope-specific CD8(+) T cells correlated with both interferon-stimulated gene levels in T cells and hepatic fibrosis score. Together, these data suggest that, compared with HCV-specific CD8(+) T cells, apoptotic epitope-specific CD8(+) T cells can better sustain chronic immune activation, owing to their capacity to produce tumor necrosis factor α, and exhibit greater resistance to inhibitory signals during chronic HCV infection.


Asunto(s)
Apoptosis , Linfocitos T CD8-positivos/inmunología , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/patología , Interferones/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Adulto , Anciano , Femenino , Humanos , Interleucina-2/metabolismo , Cirrosis Hepática/patología , Masculino , Persona de Mediana Edad , Factor de Necrosis Tumoral alfa/metabolismo
10.
Immunol Cell Biol ; 94(9): 886-894, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27265253

RESUMEN

Growing evidences put B lymphocytes on a central stage in multiple sclerosis (MS) immunopathology. While investigating the effects of interferon-ß (IFN-ß) therapy, one of the most used first-line disease-modifying drugs for the treatment of relapsing-remitting MS, in circulating B-cell sub-populations, we found a specific and marked decrease of CD27+ memory B cells. Interestingly, memory B cells are considered a population with a great disease-driving relevance in MS and resulted to be also target of B-cell depleting therapies. In addition, Epstein-Barr virus (EBV), associated with MS etiopathogenesis, harbors in this cell type and an IFN-ß-induced reduction of the memory B-cell compartment, in turn, resulted in a decreased expression of the EBV gene latent membrane protein 2A in treated patients. We found that in vivo IFN-ß therapy specifically and highly induced apoptosis in memory B cells, in accordance with a strong increase of the apoptotic markers Annexin-V and active caspase-3, via a mechanism requiring the FAS-receptor/TACI (transmembrane activator and CAML interactor) signaling. Thus, efficacy of IFN-ß therapy in MS may rely not only on its recognized anti-inflammatory activities but also on the specific depletion of memory B cells, considered to be a pathogenic cell subset, reducing their inflammatory impact in target organs.


Asunto(s)
Apoptosis/efectos de los fármacos , Linfocitos B/inmunología , Memoria Inmunológica/efectos de los fármacos , Interferón beta/farmacología , Interferón beta/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Receptor fas/metabolismo , Adulto , Linfocitos B/efectos de los fármacos , Proteína Ligando Fas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/patología , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/patología , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Adulto Joven
11.
J Immunol ; 192(5): 2291-304, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24477914

RESUMEN

The transcriptional repressor B lymphocyte-induced maturation protein 1 (BLIMP1) is a master regulator of B and T cell differentiation. To examine the role of BLIMP1 in innate immunity, we used a conditional knockout (CKO) of Blimp1 in myeloid cells and found that Blimp1 CKO mice were protected from lethal infection induced by Listeria monocytogenes. Transcriptome analysis of Blimp1 CKO macrophages identified the murine chemokine (C-C motif) ligand 8, CCL8, as a direct target of Blimp1-mediated transcriptional repression in these cells. BLIMP1-deficient macrophages expressed elevated levels of Ccl8, and consequently Blimp1 CKO mice had higher levels of circulating CCL8, resulting in increased neutrophils in the peripheral blood, promoting a more aggressive antibacterial response. Mice lacking the Ccl8 gene were more susceptible to L. monocytogenes infection than were wild-type mice. Although CCL8 failed to recruit neutrophils directly, it was chemotactic for γ/δ T cells, and CCL8-responsive γ/δ T cells were enriched for IL-17F. Finally, CCL8-mediated enhanced clearance of L. monocytogenes was dependent on γ/δ T cells. Collectively, these data reveal an important role for BLIMP1 in modulating host defenses by suppressing expression of the chemokine CCL8.


Asunto(s)
Quimiocina CCL8/inmunología , Regulación de la Expresión Génica/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Macrófagos/inmunología , Factores de Transcripción/inmunología , Animales , Quimiocina CCL8/genética , Regulación de la Expresión Génica/genética , Listeriosis/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Factores de Transcripción/genética , Transcripción Genética/genética , Transcripción Genética/inmunología
12.
PLoS Pathog ; 9(4): e1003220, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23592979

RESUMEN

It has long been known that multiple sclerosis (MS) is associated with an increased Epstein-Barr virus (EBV) seroprevalence and high immune reactivity to EBV and that infectious mononucleosis increases MS risk. This evidence led to postulate that EBV infection plays a role in MS etiopathogenesis, although the mechanisms are debated. This study was designed to assess the prevalence and magnitude of CD8+ T-cell responses to EBV latent (EBNA-3A, LMP-2A) and lytic (BZLF-1, BMLF-1) antigens in relapsing-remitting MS patients (n = 113) and healthy donors (HD) (n = 43) and to investigate whether the EBV-specific CD8+ T cell response correlates with disease activity, as defined by clinical evaluation and gadolinium-enhanced magnetic resonance imaging. Using HLA class I pentamers, lytic antigen-specific CD8+ T cell responses were detected in fewer untreated inactive MS patients than in active MS patients and HD while the frequency of CD8+ T cells specific for EBV lytic and latent antigens was higher in active and inactive MS patients, respectively. In contrast, the CD8+ T cell response to cytomegalovirus did not differ between HD and MS patients, irrespective of the disease phase. Marked differences in the prevalence of EBV-specific CD8+ T cell responses were observed in patients treated with interferon-ß and natalizumab, two licensed drugs for relapsing-remitting MS. Longitudinal studies revealed expansion of CD8+ T cells specific for EBV lytic antigens during active disease in untreated MS patients but not in relapse-free, natalizumab-treated patients. Analysis of post-mortem MS brain samples showed expression of the EBV lytic protein BZLF-1 and interactions between cytotoxic CD8+ T cells and EBV lytically infected plasma cells in inflammatory white matter lesions and meninges. We therefore propose that inability to control EBV infection during inactive MS could set the stage for intracerebral viral reactivation and disease relapse.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Herpesvirus Humano 4/inmunología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/virología , Transactivadores/metabolismo , Adulto , Anticuerpos Monoclonales Humanizados/uso terapéutico , Encéfalo/virología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Humanos , Interferón beta/uso terapéutico , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Natalizumab , Estudios Seroepidemiológicos , Adulto Joven
13.
Eur J Immunol ; 43(1): 147-58, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22996354

RESUMEN

Plasmacytoid DCs (pDCs) are crucial mediators in the establishment of immunity against most viruses, given their extraordinary capacity to produce a massive quantity of type I IFN. In this study we investigate the response of pDCs to infection with EBV, a γ-herpes virus that persists with an asymptomatic infection in immunocompetent hosts, although in certain conditions it can promote development of cancers or autoimmune diseases. We show that high amounts of type I IFNs were released from isolated pDCs after exposure to EBV by a mechanism requiring TLRs and a functional autophagic machinery. We next demonstrate that EBV can infect pDCs via viral binding to MHC class II molecule HLA-DR and that pDCs express EBV-induced latency genes. Furthermore, we observe that EBV is able to induce activation but not maturation of pDCs, which correlates with an impaired TNF-α release. Accordingly, EBV-infected pDCs are unable to mount a full T-cell response, suggesting that impaired pDC maturation, combined with a concomitant EBV-mediated upregulation of the T-cell inhibitory molecules B7-H1 and ICOS-L, could represent an immune-evasion strategy promoted by the virus. These mechanisms might lead to persistence in immunocompetent hosts or to dysregulated immune responses linked to EBV-associated diseases.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/fisiología , Linfocitos T/inmunología , Receptor Toll-Like 9/inmunología , Autofagia/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Células Cultivadas , Células Dendríticas/virología , Antígenos HLA-DR/metabolismo , Herpesvirus Humano 4/patogenicidad , Humanos , Evasión Inmune , Ligando Coestimulador de Linfocitos T Inducibles/genética , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Activación de Linfocitos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Internalización del Virus , Latencia del Virus/genética , Replicación Viral
14.
Eur J Immunol ; 43(7): 1963-72, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23636665

RESUMEN

The implication of B lymphocytes in the immunopathology of multiple sclerosis (MS) is increasingly recognized. Here we investigated the response of B cells to IFN-ß, a first-line therapy for relapsing-remitting MS patients, upon stimulation with TLR. IFN-ß restored the frequency of TLR7-induced IgM and IgG-secreting cells in MS patients to the levels found in healthy donors, showing a specific deficiency in the TLR7 pathway. However, no difference was observed in the TLR9 response. Furthermore, in MS-derived PBMCs, TLR7-mediated production of IL-6 and the ex vivo expression of B-cell-activating factor of the TNF family, two crucial cytokines for B-cell differentiation and survival, were induced by IFN-ß. Depletion of monocytes, which are key producers of both IL-6 and B-cell-activating factor of the TNF family, showed that TLR7-mediated B-cell differentiation into Ig-secreting cells is strongly dependent on the cross-talk between B cells and monocytes. Accordingly, impaired expression of TLR7 mRNA was observed in PBMCs and monocytes isolated from MS-affected individuals as compared with those from healthy donors, which was rescued by IFN-ß therapy. Collectively, our data unveil a novel TLR7-regulated mechanism in in vivo IFN-ß-stimulated whole leukocytes that could be exploited to define new TLR7-based strategies for the treatment of MS.


Asunto(s)
Linfocitos B/efectos de los fármacos , Interferón beta/uso terapéutico , Monocitos/efectos de los fármacos , Esclerosis Múltiple Recurrente-Remitente/inmunología , Receptor Cross-Talk/efectos de los fármacos , Receptor Toll-Like 7/inmunología , Adulto , Linfocitos B/metabolismo , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Factores Inmunológicos/uso terapéutico , Masculino , Monocitos/metabolismo , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Cross-Talk/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
iScience ; 27(5): 109703, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38706870

RESUMEN

Monoclonal antibodies (mAb) targeting the SARS-CoV-2 Spike (S) glycoprotein have been exploited for the treatment of severe COVID-19. In this study, we evaluated the immune-regulatory features of two neutralizing anti-S mAbs (nAbs), named J08 and F05, with wild-type (WT) conformation or silenced Fc functions. In the presence of D614G SARS-CoV-2, WT nAbs enhance intracellular viral uptake in immune cells and amplify antiviral type I Interferon and inflammatory cytokine and chemokine production without viral replication, promoting the differentiation of CD16+ inflammatory monocytes and innate/adaptive PD-L1+ and PD-L1+CD80+ plasmacytoid Dendritic Cells. In spite of a reduced neutralizing property, WT J08 nAb still promotes the IL-6 production and differentiation of CD16+ monocytes once binding Omicron BA.1 variant. Fc-mediated regulation of antiviral and inflammatory responses, in the absence of viral replication, highlighted in this study, might positively tune immune response during SARS-CoV-2 infection and be exploited also in mAb-based therapeutic and prophylactic strategies against viral infections.

16.
Ann Rheum Dis ; 72(9): 1559-68, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23268369

RESUMEN

OBJECTIVES: Rheumatoid arthritis (RA) is associated with an increased Epstein-Barr virus (EBV) blood DNA load, a robust immune response to EBV and cross-reactive circulating antibodies to viral and self-antigens. However, the role of EBV in RA pathogenesis remains elusive. Here, we investigated the relationship between synovial EBV infection, ectopic lymphoid structures (ELS) and immunity to citrullinated self and EBV proteins. METHODS: Latent and lytic EBV infection was investigated in 43 RA synovial tissues characterised for presence/absence of ELS and in 11 control osteoarthritis synovia using RT-PCR, in situ hybridisation and immunohistochemistry. Synovial production of anti-citrullinated protein (ACPA) and anti-citrullinated EBV peptide (VCP1/VCP2) antibodies was investigated in situ and in vivo in the severe combined immunodeficiency (SCID)/RA chimeric model. RESULTS: EBV dysregulation was observed exclusively in ELS+ RA but not osteoarthritis (OA) synovia, as revealed by presence of EBV latent (LMP2A, EBV-encoded small RNA (EBER)) transcripts, EBER+ cells and immunoreactivity for EBV latent (LMP1, LMP2A) and lytic (BFRF1) antigens in ELS-associated B cells and plasma cells, respectively. Importantly, a large proportion of ACPA-producing plasma cells surrounding synovial germinal centres were infected with EBV. Furthermore, ELS-containing RA synovia transplanted into SCID mice supported production of ACPA and anti-VCP1/VCP2 antibodies. Analysis of CD4+ and CD8+ T-cell localisation and granzyme B expression suggests that EBV persistence in ELS-containing synovia may be favoured by exclusion of CD8+ T cells from B-cell follicles and impaired CD8-mediated cytotoxicity. CONCLUSIONS: We demonstrated active EBV infection within ELS in the RA synovium in association with local differentiation of ACPA-reactive B cells.


Asunto(s)
Artritis Reumatoide/virología , Autoinmunidad , Herpesvirus Humano 4/fisiología , Osteoartritis/virología , Células Plasmáticas/virología , Membrana Sinovial/virología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Femenino , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Tejido Linfoide , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Osteoartritis/inmunología , Osteoartritis/patología , Células Plasmáticas/inmunología , Células Plasmáticas/patología , Membrana Sinovial/inmunología , Membrana Sinovial/patología , Carga Viral
17.
Int Immunopharmacol ; 117: 109996, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36933449

RESUMEN

The peculiar property of Thymosin alpha 1 (Tα1) to act as master regulator of immune homeostasis has been successfully defined in different physiological and pathological contexts ranging from cancer to infection. Interestingly, recent papers also demonstrated its mitigating effect on the "cytokine storm" as well as on the T-cell exhaustion/activation in SARS-CoV-2 infected individuals. Nevertheless, in spite of the increasing knowledge on Tα1-induced effects on T cell response confirming the distinctive features of this multifaceted peptide, little is known on its effects on innate immunity during SARS-CoV-2 infection. Here, we interrogated peripheral blood mononuclear cell (PBMC) cultures stimulated with SARS-CoV-2 to disclose Tα1 properties on the main cell players of early response to infection, namely monocytes and myeloid dendritic cells (mDC). Moving from ex vivo data showing an enhancement in the frequency of inflammatory monocytes and activated mDC in COVID-19 patients, a PBMC-based experimental setting reproduced in vitro a similar profile with an increased percentage of CD16+ inflammatory monocytes and mDC expressing CD86 and HLA-DR activation markers in response to SARS-CoV-2 stimulation. Interestingly, the treatment of SARS-CoV-2-stimulated PBMC with Tα1 dampened the inflammatory/activation status of both monocytes and mDC by reducing the release of pro-inflammatory mediators, including TNF-α, IL-6 and IL-8, while promoting the production of the anti-inflammatory cytokine IL-10. This study further clarifies the working hypothesis on Tα1 mitigating action on COVID-19 inflammatory condition. Moreover, these evidence shed light on inflammatory pathways and cell types involved in acute SARS-CoV-2 infection and likely targetable by newly immune-regulating therapeutic approaches.


Asunto(s)
COVID-19 , Timosina , Humanos , Timalfasina/uso terapéutico , Leucocitos Mononucleares/metabolismo , SARS-CoV-2/metabolismo , Citocinas/metabolismo , Inflamación/tratamiento farmacológico , Timosina/farmacología , Timosina/uso terapéutico
18.
Clin Transl Immunology ; 12(3): e1434, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36969367

RESUMEN

Objectives: The very rapidly approved mRNA-based vaccines against SARS-CoV-2 spike glycoprotein, including Pfizer-BioNTech BNT162b2, are effective in protecting from severe coronavirus disease 2019 (COVID-19) in immunocompetent population. However, establishing the duration and identifying correlates of vaccine-induced protection will be crucial to optimise future immunisation strategies. Here, we studied in healthy vaccine recipients and people with multiple sclerosis (pwMS), undergoing different therapies, the regulation of innate immune response by mRNA vaccination in order to correlate it with the magnitude of vaccine-induced protective humoral responses. Methods: Healthy subjects (n = 20) and matched pwMS (n = 22) were longitudinally sampled before and after mRNA vaccination. Peripheral blood mononuclear cell (PBMC)-associated type I and II interferon (IFN)-inducible gene expression, serum innate cytokine/chemokine profile as well as binding and neutralising anti-SARS-COV-2 antibodies (Abs) were measured. Results: We identified an early immune module composed of the IFN-inducible genes Mx1, OAS1 and IRF1, the serum cytokines IL-15, IL-6, TNF-α and IFN-γ and the chemokines IP-10, MCP-1 and MIG, induced 1 day post second and third BNT162b2 vaccine doses, strongly correlating with magnitude of humoral response to vaccination in healthy and MS vaccinees. Moreover, induction of the early immune module was dramatically affected in pwMS treated with fingolimod and ocrelizumab, both groups unable to induce a protective humoral response to COVID-19 vaccine. Conclusion: Overall, this study suggests that the vaccine-induced early regulation of innate immunity is mediated by IFN signalling, impacts on the magnitude of adaptive responses and it might be indicative of vaccine-induced humoral protection.

19.
Immunol Cell Biol ; 89(3): 437-46, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20805841

RESUMEN

Mycobacterium tuberculosis (Mtb) evades the immune response by impairing the functions of different antigen-presenting cells. We have recently shown that Mtb hijacks differentiation of monocytes into dendritic cells (DCs). To further characterize the mechanisms underlying this process, we investigated the consequences of inducing dendritic cell differentiation using interferon-α and granulocyte-macrophage colony-stimulating factor in the presence of supernatants (SNs) obtained from monocyte cultures treated with or without heat-inactivated Mtb. Although the SNs from control cultures do not interfere with the generation of fully differentiated DCs, monocytes stimulated with SNs from Mtb-stimulated cells (SN Mtb) remained CD14(+) and poorly differentiated into CD1a(+) cells. Among cytokines known to affect dendritic cell differentiation, we observed a robust production of interleukin-1ß, interleukin-6, interleukin-10 and tumor necrosis factor-α upon Mtb stimulation. However, only interleukin-10 neutralization through the addition of soluble interleukin-10 receptor reversed the inhibitory activity of SN Mtb. Accordingly, the addition of recombinant interleukin-10 was able to significantly reduce CD1a expression. The interaction of Mtb with differentiating monocytes rapidly activates p38 mitogen-activated protein kinase, signal transducer and activator of transcription pathways, which are likely involved in interleukin-10 gene expression. Taken together, our results suggest that Mtb may inhibit the differentiation of bystander non-infected monocytes into DCs through the release of interleukin-10. These results shed light on new aspects of the host-pathogen interaction, which might help to identify innovative immunological strategies to limit Mtb virulence.


Asunto(s)
Efecto Espectador , Diferenciación Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Interleucina-10/inmunología , Mycobacterium tuberculosis/fisiología , Tuberculosis/inmunología , Efecto Espectador/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Citocinas/biosíntesis , Humanos , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Mycobacterium tuberculosis/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Virol ; 84(20): 10802-11, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20686034

RESUMEN

The discovery of the Toll-like receptors (TLRs) and their importance in the regulation of host responses to infection raised attention to the complex interplay between viral gene products and the host innate immune responses in determining the outcome of virus infection. Robust inflammatory cytokine responses are observed in herpes simplex virus (HSV)-infected animals and cells. Our studies have demonstrated that Toll-like receptor 2 (TLR2) activation by HSV results in NF-κB activation with concomitant inflammatory cytokine production and that TLR2 activation plays a critical role in HSV-induced pathology and mortality. Here we demonstrate that the HSV-1 immediate-early ICP0 protein reduces the TLR2-mediated inflammatory response to HSV 1 (HSV-1) infection. Expression of ICP0 alone is sufficient to block TLR2-driven responses to both viral and nonviral ligands at or downstream of the MyD88 adaptor and upstream of p65. ICP0 alone can also reduce the levels of MyD88 and Mal (TIRAP). In HSV-infected cells, the E3 ligase function of ICP0 and cellular proteasomal activity are required for the inhibitory activity. Our results argue for a model in which ICP0 promotes the degradation of TLR adaptor molecules and inhibition of the inflammatory response, much as it inhibits the interferon response by sequestration and degradation of interferon regulatory factor 3 (IRF-3).


Asunto(s)
Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/fisiología , FN-kappa B/fisiología , Receptor Toll-Like 2/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/fisiología , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Citocinas/biosíntesis , Genes Virales , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidad , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Humanos , Proteínas Inmediatas-Precoces/genética , Mediadores de Inflamación/fisiología , Proteínas de Transporte de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación , Proteínas de la Mielina/fisiología , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito , Factor 88 de Diferenciación Mieloide/fisiología , Proteolípidos/fisiología , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/fisiología , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA