Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(5): 1282-1297.e18, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33098771

RESUMEN

Classically considered short-lived and purely defensive leukocytes, neutrophils are unique in their fast and moldable response to stimulation. This plastic behavior may underlie variable and even antagonistic functions during inflammation or cancer, yet the full spectrum of neutrophil properties as they enter healthy tissues remains unexplored. Using a new model to track neutrophil fates, we found short but variable lifetimes across multiple tissues. Through analysis of the receptor, transcriptional, and chromatin accessibility landscapes, we identify varying neutrophil states and assign non-canonical functions, including vascular repair and hematopoietic homeostasis. Accordingly, depletion of neutrophils compromised angiogenesis during early age, genotoxic injury, and viral infection, and impaired hematopoietic recovery after irradiation. Neutrophils acquired these properties in target tissues, a process that, in the lungs, occurred in CXCL12-rich areas and relied on CXCR4. Our results reveal that tissues co-opt neutrophils en route for elimination to induce programs that support their physiological demands.


Asunto(s)
Linaje de la Célula , Neutrófilos/metabolismo , Especificidad de Órganos , Animales , Cromatina/metabolismo , Femenino , Hematopoyesis , Intestinos/irrigación sanguínea , Pulmón/irrigación sanguínea , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores CXCR4/metabolismo , Análisis de la Célula Individual , Transcripción Genética , Transcriptoma/genética
2.
Nat Immunol ; 17(9): 1037-1045, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27348412

RESUMEN

Macrophages tightly scale their core metabolism after being activated, but the precise regulation of the mitochondrial electron-transport chain (ETC) and its functional implications are currently unknown. Here we found that recognition of live bacteria by macrophages transiently decreased assembly of the ETC complex I (CI) and CI-containing super-complexes and switched the relative contributions of CI and CII to mitochondrial respiration. This was mediated by phagosomal NADPH oxidase and the reactive oxygen species (ROS)-dependent tyrosine kinase Fgr. It required Toll-like receptor signaling and the NLRP3 inflammasome, which were both connected to bacterial viability-specific immune responses. Inhibition of CII during infection with Escherichia coli normalized serum concentrations of interleukin 1ß (IL-1ß) and IL-10 to those in mice treated with dead bacteria and impaired control of bacteria. We have thus identified ETC adaptations as an early immunological-metabolic checkpoint that adjusts innate immune responses to bacterial infection.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Infecciones por Escherichia coli/inmunología , Escherichia coli K12/inmunología , Macrófagos/inmunología , Mitocondrias/metabolismo , Animales , Células Cultivadas , Metabolismo Energético/genética , Interacciones Huésped-Parásitos , Inmunidad Innata/genética , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fagocitosis , Especies Reactivas de Oxígeno/metabolismo
3.
Int J Mol Sci ; 24(10)2023 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-37240378

RESUMEN

The stimulator of interferon genes (STING) is an adaptor protein involved in the activation of IFN-ß and many other genes associated with the immune response activation in vertebrates. STING induction has gained attention from different angles such as the potential to trigger an early immune response against different signs of infection and cell damage, or to be used as an adjuvant in cancer immune treatments. Pharmacological control of aberrant STING activation can be used to mitigate the pathology of some autoimmune diseases. The STING structure has a well-defined ligand binding site that can harbor natural ligands such as specific purine cyclic di-nucleotides (CDN). In addition to a canonical stimulation by CDNs, other non-canonical stimuli have also been described, whose exact mechanism has not been well defined. Understanding the molecular insights underlying the activation of STING is important to realize the different angles that need to be considered when designing new STING-binding molecules as therapeutic drugs since STING acts as a versatile platform for immune modulators. This review analyzes the different determinants of STING regulation from the structural, molecular, and cell biology points of view.


Asunto(s)
Adyuvantes Inmunológicos , Nucleótidos Cíclicos , Animales , Sitios de Unión
4.
Int J Mol Sci ; 23(19)2022 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-36232593

RESUMEN

Type I interferons (IFN), including IFNß, play a protective role in multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Type I IFNs are induced by the stimulation of innate signaling, including via cytoplasmic RIG-I-like receptors. In the present study, we investigated the potential effect of a chimeric protein containing the key domain of RIG-I signaling in the production of CNS endogenous IFNß and asked whether this would exert a therapeutic effect against EAE. We intrathecally administered an adeno-associated virus vector (AAV) encoding a fusion protein comprising RIG-I 2CARD domains (C) and the first 200 amino acids of mitochondrial antiviral-signaling protein (MAVS) (M) (AAV-CM). In vivo imaging in IFNß/luciferase reporter mice revealed that a single intrathecal injection of AAV-CM resulted in dose-dependent and sustained IFNß expression within the CNS. IFNß expression was significantly increased for 7 days. Immunofluorescent staining in IFNß-YFP reporter mice revealed extraparenchymal CD45+ cells, choroid plexus, and astrocytes as sources of IFNß. Moreover, intrathecal administration of AAV-CM at the onset of EAE induced the suppression of EAE, which was IFN-I-dependent. These findings suggest that accessing the signaling pathway downstream of RIG-I represents a promising therapeutic strategy for inflammatory CNS diseases, such as MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Interferón Tipo I , Aminoácidos , Animales , Antivirales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Interferón Tipo I/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Ratones , Proteínas Recombinantes de Fusión , Transducción de Señal
5.
Int J Mol Sci ; 21(20)2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33066689

RESUMEN

Glioma tumors are one of the most devastating cancer types. Glioblastoma is the most advanced stage with the worst prognosis. Current therapies are still unable to provide an effective cure. Recent advances in oncolytic immunotherapy have generated great expectations in the cancer therapy field. The use of oncolytic viruses (OVs) in cancer treatment is one such immune-related therapeutic alternative. OVs have a double oncolytic action by both directly destroying the cancer cells and stimulating a tumor specific immune response to return the ability of tumors to escape the control of the immune system. OVs are one promising alternative to conventional therapies in glioma tumor treatment. Several clinical trials have proven the feasibility of using some viruses to specifically infect tumors, eluding undesired toxic effects in the patient. Here, we revisited the literature to describe the main OVs proposed up to the present moment as therapeutic alternatives in order to destroy glioma cells in vitro and trigger tumor destruction in vivo. Oncolytic viruses were divided with respect to the genome in DNA and RNA viruses. Here, we highlight the results obtained in various clinical trials, which are exploring the use of these agents as an alternative where other approaches provide limited hope.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Viroterapia Oncolítica/métodos , Animales , Ensayos Clínicos como Asunto , Humanos , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Virus Oncolíticos/fisiología
7.
Mol Cancer ; 14: 210, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26671477

RESUMEN

BACKGROUND: The limited efficacy of current treatments against pancreatic cancer has prompted the search of new alternatives such as virotherapy. Activation of the immune response against cancer cells is emerging as one of the main mechanisms of action of oncolytic viruses (OV). Direct oncolysis releases tumor antigens, and viral replication within the tumor microenvironment is a potent danger signal. Arming OV with immunostimulatory transgenes further enhances their therapeutic effect. However, standard virotherapy protocols do not take full advantage of OV as cancer vaccines because repeated viral administrations may polarize immune responses against strong viral antigens, and the rapid onset of neutralizing antibodies limits the efficacy of redosing. An alternative paradigm based on sequential combination of antigenically distinct OV has been recently proposed. METHODS: We have developed a protocol consisting of sequential intratumor administrations of new Adenovirus (Ad) and Newcastle Disease Virus (NDV)-based OV encoding the immunostimulatory cytokine oncostatin M (OSM). Transgene expression, toxicity and antitumor effect were evaluated using an aggressive orthotopic pancreatic cancer model in Syrian hamsters, which are sensitive to OSM and permissive for replication of both OVs. RESULTS: NDV-OSM was more cytolytic, whereas Ad-OSM caused higher OSM expression in vivo. Both viruses achieved only a marginal antitumor effect in monotherapy. In addition, strong secretion of OSM in serum limited the maximal tolerated dose of Ad-OSM. In contrast, moderate doses of Ad-OSM followed one week later by NDV-OSM were safe, showed a significant antitumor effect and stimulated immune responses against cancer cells. Similar efficacy was observed when the order of virus administrations was reversed. CONCLUSION: Sequential administration of oncolytic Ad and NDV encoding OSM is a promising approach against pancreatic cancer.


Asunto(s)
Viroterapia Oncolítica/métodos , Oncostatina M/biosíntesis , Neoplasias Pancreáticas/terapia , Animales , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Cricetinae , Humanos , Mesocricetus , Trasplante de Neoplasias , Virus Oncolíticos/genética , Oncostatina M/genética , Replicación Viral
8.
J Immunol ; 190(12): 6694-706, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23690480

RESUMEN

Agonist anti-CD137 (4-1BB) mAbs enhance CD8-mediated antitumor immunity. Agonist anti-human CD137 mAbs binding to four distinct epitopes on the CD137 glycoprotein costimulated T cell activation irrespective of the engaged epitope or its interference with CD137L binding. CD137 perturbation with all these agonist mAbs resulted in Ag and Ab internalization toward an endosomal vesicular compartment. Internalization was observed in activated T lymphocytes from humans and mice, not only in culture but also in Ab-injected living animals. These in vivo experiments were carried out upon systemic i.v. injections with anti-CD137 mAbs and showed CD137 internalization in tumor-infiltrating lymphocytes and in activated human T cells transferred to immunodeficient mice. Efficient CD137 internalization required K63 polyubiquitination and endocytosed CD137-containing vesicles recruited TNFR-associated factor (TRAF) 2 and were decorated with K63 polyubiquitins. CD137 stimulation activates NF-κB through a K63-linked polyubiquitination-dependent route, and CD137-associated TRAF2 becomes K63 polyubiquitinated. Consistent with a role for TRAF2 in CD137 signaling, transgenic mice functionally deficient in TRAF2 showed delayed immunotherapeutic activity of anti-CD137 mAbs. As a whole, these findings advance our knowledge of the mechanisms of action of anti-CD137 immunostimulatory mAbs such as those currently undergoing clinical trials in cancer patients.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Activación de Linfocitos/inmunología , Neoplasias Experimentales/inmunología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Western Blotting , Línea Celular , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Endosomas/efectos de los fármacos , Endosomas/inmunología , Endosomas/metabolismo , Femenino , Humanos , Inmunoprecipitación , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/inmunología , FN-kappa B/metabolismo , Neoplasias Experimentales/terapia , Poliubiquitina/inmunología , Poliubiquitina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factor 2 Asociado a Receptor de TNF/inmunología , Factor 2 Asociado a Receptor de TNF/metabolismo , Transfección , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
9.
Int J Cancer ; 135(11): 2516-27, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24473991

RESUMEN

New mouse models with specific drivers of genetic alterations are needed for preclinical studies. Herein, we created and characterized at the genetic level a new syngeneic model for lung cancer and metastasis in Balb-c mice. Tumor cell lines were obtained from a silica-mediated airway chronic inflammation that promotes tumorigenesis when combined with low doses of N-nitrosodimethylamine, a tobacco smoke carcinogen. Orthotopic transplantation of these cells induced lung adenocarcinomas, and their intracardiac injection led to prominent colonization of various organs (bone, lung, liver and brain). Driver gene alterations included a mutation in the codon 12 of KRAS (G-A transition), accompanied by a homozygous deletion of the WW domain-containing oxidoreductase (WWOX) gene. The mutant form of WWOX lacked exons 5-8 and displayed reduced protein expression level and activity. WWOX gene restoration decreased the in vitro and in vivo tumorigenicity, confirming the tumor suppressor function of this gene in this particular model. Interestingly, we found that cells displayed remarkable sphere formation ability with expression of specific lung cancer stem cell markers. Study of non-small-cell lung cancer patient cohorts demonstrated a deletion of WWOX in 30% of cases, with significant reduction in protein levels as compared to normal tissues. Overall, our new syngeneic mouse model provides a most valuable tool to study lung cancer metastasis in balb-c mice background and highlights the importance of WWOX deletion in lung carcinogenesis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/secundario , Modelos Animales de Enfermedad , Inflamación/patología , Neoplasias Pulmonares/patología , Recurrencia Local de Neoplasia/patología , Oxidorreductasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Supresoras de Tumor/genética , Proteínas ras/genética , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Animales , Apoptosis , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Proliferación Celular , Hibridación Genómica Comparativa , Transición Epitelial-Mesenquimal , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Inflamación/genética , Inflamación/mortalidad , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación/genética , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Estadificación de Neoplasias , Oxidorreductasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo , Oxidorreductasa que Contiene Dominios WW , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/metabolismo
10.
PLoS Pathog ; 8(11): e1003059, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209422

RESUMEN

Influenza A viruses can adapt to new host species, leading to the emergence of novel pathogenic strains. There is evidence that highly pathogenic viruses encode for non-structural 1 (NS1) proteins that are more efficient in suppressing the host immune response. The NS1 protein inhibits type-I interferon (IFN) production partly by blocking the TRIM25 ubiquitin E3 ligase-mediated Lys63-linked ubiquitination of the viral RNA sensor RIG-I, required for its optimal downstream signaling. In order to understand possible mechanisms of viral adaptation and host tropism, we examined the ability of NS1 encoded by human (Cal04), avian (HK156), swine (SwTx98) and mouse-adapted (PR8) influenza viruses to interact with TRIM25 orthologues from mammalian and avian species. Using co-immunoprecipitation assays we show that human TRIM25 binds to all tested NS1 proteins, whereas the chicken TRIM25 ortholog binds preferentially to the NS1 from the avian virus. Strikingly, none of the NS1 proteins were able to bind mouse TRIM25. Since NS1 can inhibit IFN production in mouse, we tested the impact of TRIM25 and NS1 on RIG-I ubiquitination in mouse cells. While NS1 efficiently suppressed human TRIM25-dependent ubiquitination of RIG-I 2CARD, NS1 inhibited the ubiquitination of full-length mouse RIG-I in a mouse TRIM25-independent manner. Therefore, we tested if the ubiquitin E3 ligase Riplet, which has also been shown to ubiquitinate RIG-I, interacts with NS1. We found that NS1 binds mouse Riplet and inhibits its activity to induce IFN-ß in murine cells. Furthermore, NS1 proteins of human but not swine or avian viruses were able to interact with human Riplet, thereby suppressing RIG-I ubiquitination. In conclusion, our results indicate that influenza NS1 protein targets TRIM25 and Riplet ubiquitin E3 ligases in a species-specific manner for the inhibition of RIG-I ubiquitination and antiviral IFN production.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Interferones/biosíntesis , Ubiquitinación , Proteínas no Estructurales Virales/metabolismo , Animales , Chlorocebus aethiops , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Perros , Células HeLa , Humanos , Virus de la Influenza A/genética , Gripe Humana/genética , Interferones/genética , Ratones , Ratones Noqueados , Receptores Inmunológicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Células Vero , Proteínas no Estructurales Virales/genética
11.
Microbiol Spectr ; 12(1): e0363522, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38051056

RESUMEN

IMPORTANCE: Influenza virus infection affects both lung and intestinal bacterial community composition. Most of the published analyses focus on the characterization of the microbiota composition changes. Here we assess functional alterations of gut microbiota such as nutrient and antibiotic resistance changes during an acute respiratory tract infection. Upon influenza A virus (IAV) infection, cecal microbiota drops accompanied by a decrease in the ability to metabolize some common nutrients under aerobic conditions. At the same time, the cecal community presents an increase in resistance against clinically relevant antibiotics, particularly cephalosporins. Functional characterization of complex communities presents an additional and necessary element of analysis that nowadays is mainly limited to taxonomic description. The consequences of these functional alterations could affect treatment strategies, especially in multimicrobial infections.


Asunto(s)
Microbioma Gastrointestinal , Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Humanos , Gripe Humana/tratamiento farmacológico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico
12.
Int Rev Cell Mol Biol ; 379: 221-239, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37541725

RESUMEN

Lung tumors are one of the most aggressive threats affecting humans. Current therapeutic approaches have improved patients' survival; however, further efforts are required to increase effectiveness and protection against tumor relapse and metastasis. Immunotherapy presents an alternative to previous treatments that focuses on stimulating of the patient's immune system to destroy tumor cells. Viruses can be used as part of the immune therapeutic approach as agents that could selectively infect tumor cells, triggering an immune response against the infection and against the tumor cells. Some viruses have been selected for specifically infecting and destroying cancer cells, activating the immune response, enhancing access, amplifying the cytotoxicity against the tumor cells, and improving the long-term memory that can prevent tumor relapse. Oncolytic virotherapy can then be used as a strategy to target the destruction of transformed cells at the tumor site and act in locations distant from the primary targeted tumor site. Some of the current challenges in lung cancer treatment can be addressed using traditional therapies combined with oncolytic virotherapy. Defining the best combination, including the choice of the right settings will be at the next frontier in lung cancer treatment.


Asunto(s)
Neoplasias Pulmonares , Neoplasias , Viroterapia Oncolítica , Virus , Humanos , Recurrencia Local de Neoplasia/terapia , Neoplasias/terapia , Neoplasias Pulmonares/terapia , Inmunoterapia
13.
Vaccines (Basel) ; 11(10)2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37897020

RESUMEN

Alongside the development and progress in cancer immunotherapy, research in oncolytic viruses (OVs) continues advancing novel treatment strategies to the clinic. With almost 50 clinical trials carried out over the last decade, the opportunities for intervention using OVs are expanding beyond the old-fashioned concept of "lytic killers", with promising breakthrough therapeutic strategies focused on leveraging the immunostimulatory potential of different viral platforms. This review presents an overview of non-human-adapted RNA viruses engineered for cancer therapy. Moreover, we describe the diverse strategies employed to manipulate the genomes of these viruses to optimize their therapeutic capabilities. By focusing on different aspects of this particular group of viruses, we describe the insights into the promising advancements in the field of virotherapy and its potential to revolutionize cancer treatment.

14.
Int J Infect Dis ; 131: 173-179, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37030656

RESUMEN

BACKGROUND: The clinical burden of influenza is increasing worldwide. Aging, immunosuppression, and underlying respiratory illness are determinants of poor clinical outcomes, including greater mortality. Bacterial infections seem to be the main reason. Updated information on the role of bacterial infection as the cause of complications would be of value in improving the prognosis of patients with influenza. METHODS: A systematic review and meta-analysis were performed by using the PubMed repository using keywords like: Influenza, H1N1, Streptococcus pneumoniae, bacterial coinfection, secondary coinfection, bacterial complications in pneumonia, and seasonal influenza. Only articles written in English were included in publications from 2010 to 2020. The analyses were conducted following the preferred reporting items for systematic review and meta-analyses guidelines. The results were independently validated using a TrinetX database cohort of roughly 4 million patients. RESULTS: We included 135 studies that contained data from 48,259 patients hospitalized with influenza of any age. Bacterial infections were diagnosed in 5391 (11.2%). Streptococcus pneumoniae (30.7%) and Staphylococcus aureus (30.4%) were the most frequent microorganisms, followed by Haemophilus influenzae (7.1%) and Pseudomonas aeruginosa (5.9%). The random-effects model of the meta-analysis indicated that bacterial infections posed a 3.4-fold increased risk of death compared with influenza infection alone. Unexpectedly, asthma was protective (odds ratio 0.8). CONCLUSION: Bacterial infections diagnosed in 11.2% of patients with influenza increase 3.4-fold the mortality risk. S. pneumoniae, S. aureus, H. influenzae, and P. aeruginosa account for nearly 75% of the cases. Earlier diagnosis and use of antibiotics should improve outcomes in this population.


Asunto(s)
Coinfección , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Neumonía , Infecciones Estafilocócicas , Humanos , Gripe Humana/complicaciones , Gripe Humana/tratamiento farmacológico , Gripe Humana/diagnóstico , Staphylococcus aureus , Coinfección/epidemiología , Neumonía/epidemiología , Streptococcus pneumoniae , Infecciones Estafilocócicas/epidemiología , Haemophilus influenzae
15.
Nat Commun ; 14(1): 6772, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880206

RESUMEN

Antigen cognate dendritic cell (DC)-T cell synaptic interactions drive activation of T cells and instruct DCs. Upon receiving CD4+ T cell help, post-synaptic DCs (psDCs) are licensed to generate CD8+ T cell responses. However, the cellular and molecular mechanisms that enable psDCs licensing remain unclear. Here, we describe that antigen presentation induces an upregulation of MHC-I protein molecules and increased lipid peroxidation on psDCs in vitro and in vivo. We also show that these events mediate DC licensing. In addition, psDC adoptive transfer enhances pathogen-specific CD8+ T responses and protects mice from infection in a CD8+ T cell-dependent manner. Conversely, depletion of psDCs in vivo abrogates antigen-specific CD8+ T cell responses during immunization. Together, our data show that psDCs enable CD8+ T cell responses in vivo during vaccination and reveal crucial molecular events underlying psDC licensing.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Ratones , Animales , Regulación hacia Arriba , Peroxidación de Lípido , Presentación de Antígeno , Antígenos , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Dendríticas , Sinapsis/metabolismo , Ratones Endogámicos C57BL
16.
Front Immunol ; 14: 1156603, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37143685

RESUMEN

Background: Managing the inflammatory response to SARS-Cov-2 could prevent respiratory insufficiency. Cytokine profiles could identify cases at risk of severe disease. Methods: We designed a randomized phase II clinical trial to determine whether the combination of ruxolitinib (5 mg twice a day for 7 days followed by 10 mg BID for 7 days) plus simvastatin (40 mg once a day for 14 days), could reduce the incidence of respiratory insufficiency in COVID-19. 48 cytokines were correlated with clinical outcome. Participants: Patients admitted due to COVID-19 infection with mild disease. Results: Up to 92 were included. Mean age was 64 ± 17, and 28 (30%) were female. 11 (22%) patients in the control arm and 6 (12%) in the experimental arm reached an OSCI grade of 5 or higher (p = 0.29). Unsupervised analysis of cytokines detected two clusters (CL-1 and CL-2). CL-1 presented a higher risk of clinical deterioration vs CL-2 (13 [33%] vs 2 [6%] cases, p = 0.009) and death (5 [11%] vs 0 cases, p = 0.059). Supervised Machine Learning (ML) analysis led to a model that predicted patient deterioration 48h before occurrence with a 85% accuracy. Conclusions: Ruxolitinib plus simvastatin did not impact the outcome of COVID-19. Cytokine profiling identified patients at risk of severe COVID-19 and predicted clinical deterioration. Trial registration: https://clinicaltrials.gov/, identifier NCT04348695.


Asunto(s)
COVID-19 , Deterioro Clínico , Insuficiencia Respiratoria , Humanos , Femenino , Masculino , SARS-CoV-2 , Resultado del Tratamiento
17.
PLoS Pathog ; 6(1): e1000745, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-20126449

RESUMEN

The recent 2009 pandemic H1N1 virus infection in humans has resulted in nearly 5,000 deaths worldwide. Early epidemiological findings indicated a low level of infection in the older population (>65 years) with the pandemic virus, and a greater susceptibility in people younger than 35 years of age, a phenomenon correlated with the presence of cross-reactive immunity in the older population. It is unclear what virus(es) might be responsible for this apparent cross-protection against the 2009 pandemic H1N1 virus. We describe a mouse lethal challenge model for the 2009 pandemic H1N1 strain, used together with a panel of inactivated H1N1 virus vaccines and hemagglutinin (HA) monoclonal antibodies to dissect the possible humoral antigenic determinants of pre-existing immunity against this virus in the human population. By hemagglutinination inhibition (HI) assays and vaccination/challenge studies, we demonstrate that the 2009 pandemic H1N1 virus is antigenically similar to human H1N1 viruses that circulated from 1918-1943 and to classical swine H1N1 viruses. Antibodies elicited against 1918-like or classical swine H1N1 vaccines completely protect C57B/6 mice from lethal challenge with the influenza A/Netherlands/602/2009 virus isolate. In contrast, contemporary H1N1 vaccines afforded only partial protection. Passive immunization with cross-reactive monoclonal antibodies (mAbs) raised against either 1918 or A/California/04/2009 HA proteins offered full protection from death. Analysis of mAb antibody escape mutants, generated by selection of 2009 H1N1 virus with these mAbs, indicate that antigenic site Sa is one of the conserved cross-protective epitopes. Our findings in mice agree with serological data showing high prevalence of 2009 H1N1 cross-reactive antibodies only in the older population, indicating that prior infection with 1918-like viruses or vaccination against the 1976 swine H1N1 virus in the USA are likely to provide protection against the 2009 pandemic H1N1 virus. This data provides a mechanistic basis for the protection seen in the older population, and emphasizes a rationale for including vaccination of the younger, naïve population. Our results also support the notion that pigs can act as an animal reservoir where influenza virus HAs become antigenically frozen for long periods of time, facilitating the generation of human pandemic viruses.


Asunto(s)
Anticuerpos Antivirales/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Animales , Reacciones Cruzadas , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Virulencia
18.
J Immunol ; 185(1): 424-32, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20511549

RESUMEN

Infection of human dendritic cells (DCs) by negative-strand RNA viruses, such as Newcastle disease virus, leads to the induction of the IFNbeta gene, IFNB1, through the activation of the RNA helicase RIG-I, which is encoded by DDX58. Expression levels of IFNB1 and DDX58 in infected DCs showed positive correlations at the population and the single-cell levels. DDX58 has a common and potentially functional single nucleotide polymorphism, rs10813831 (A/G), encoding an Arg7Cys amino acid change in the RIG-I protein caspase recruitment domain (CARD). Quantitative RT-PCR analysis on Newcastle disease virus-infected primary DCs from 130 individuals revealed a significant association of the Arg7Cys single nucleotide polymorphism with increased IFNB1 and DDX58 transcription. Allelic imbalance analysis ruled out allele-specific DDX58 message levels and suggested that the observed association between Arg7Cys and IFNB1 and DDX58 transcription originated from a functional change in RIG-I due to the amino acid substitution in the CARD. DDX58 transfection experiments in 293T cells confirmed a biological functional difference between RIG-I 7Cys and the more common RIG-I 7Arg. Taken together, these data indicate that the innate immune response to viral infection of human cells is modified by a functional polymorphism in the RIG-I CARD.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , ARN Helicasas DEAD-box/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunidad Innata/genética , Polimorfismo de Nucleótido Simple/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD/fisiología , Caspasas/genética , Línea Celular , Pollos , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/biosíntesis , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/fisiología , Células Dendríticas/virología , Humanos , Interferón beta/biosíntesis , Interferón beta/genética , Virus de la Enfermedad de Newcastle/inmunología , Estructura Terciaria de Proteína/genética , Receptores Inmunológicos , Activación Transcripcional/inmunología
19.
Mater Today Bio ; 13: 100191, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35024597

RESUMEN

Inorganic materials can provide a set of tools to decontaminate solid, liquid or air containing viral particles. The use of disinfectants can be limited or not practical in scenarios where continuous cleaning is not feasible. Physicochemical differences between viruses raise the need for effective formulations for all kind of viruses. In the present work we describe two types of antimicrobial inorganic materials: i) a novel soda-lime glass (G3), and ii) kaolin containing metals nanoparticles (Ag or CuO), as materials to disable virus infectivity. Strong antiviral properties can be observed in G3 glass, and kaolin-containing nanoparticle materials showing a reduction of viral infectivity close to 99%. in the first 10 â€‹min of contact of vesicular stomatitis virus (VSV). A potent virucidal activity is also present in G3 and kaolin containing Ag or CuO nanoparticles against all kinds of viruses tested, reducing more than 99% the amount of HSV-1, Adenovirus, VSV, Influenza virus and SARS-CoV-2 exposed to them. Virucidal properties could be explained by a direct interaction of materials with viruses as well as inactivation by the presence of virucidal elements in the material lixiviates. Kaolin-based materials guarantee a controlled release of active nanoparticles with antiviral activity. Current coronavirus crisis highlights the need for new strategies to remove viruses from contaminated areas. We propose these low-cost inorganic materials as useful disinfecting antivirals in the actual or future pandemic threats.

20.
Cell Rep ; 38(1): 110184, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986349

RESUMEN

MV130 is an inactivated polybacterial mucosal vaccine that confers protection to patients against recurrent respiratory infections, including those of viral etiology. However, its mechanism of action remains poorly understood. Here, we find that intranasal prophylaxis with MV130 modulates the lung immune landscape and provides long-term heterologous protection against viral respiratory infections in mice. Intranasal administration of MV130 provides protection against systemic candidiasis in wild-type and Rag1-deficient mice lacking functional lymphocytes, indicative of innate immune-mediated protection. Moreover, pharmacological inhibition of trained immunity with metformin abrogates the protection conferred by MV130 against influenza A virus respiratory infection. MV130 induces reprogramming of both mouse bone marrow progenitor cells and in vitro human monocytes, promoting an enhanced cytokine production that relies on a metabolic shift. Our results unveil that the mucosal administration of a fully inactivated bacterial vaccine provides protection against viral infections by a mechanism associated with the induction of trained immunity.


Asunto(s)
Vacunas Bacterianas/inmunología , Inmunidad Mucosa/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Mucosa Respiratoria/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Administración Intranasal , Animales , Anticuerpos Antivirales/inmunología , Bacterias/inmunología , Vacunas Bacterianas/administración & dosificación , Candidiasis/prevención & control , Línea Celular , Chlorocebus aethiops , Citocinas/biosíntesis , Humanos , Virus de la Influenza A/inmunología , Células L , Pulmón/inmunología , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA