Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nature ; 624(7992): 645-652, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38093014

RESUMEN

People with diabetes feature a life-risking susceptibility to respiratory viral infection, including influenza and SARS-CoV-2 (ref. 1), whose mechanism remains unknown. In acquired and genetic mouse models of diabetes, induced with an acute pulmonary viral infection, we demonstrate that hyperglycaemia leads to impaired costimulatory molecule expression, antigen transport and T cell priming in distinct lung dendritic cell (DC) subsets, driving a defective antiviral adaptive immune response, delayed viral clearance and enhanced mortality. Mechanistically, hyperglycaemia induces an altered metabolic DC circuitry characterized by increased glucose-to-acetyl-CoA shunting and downstream histone acetylation, leading to global chromatin alterations. These, in turn, drive impaired expression of key DC effectors including central antigen presentation-related genes. Either glucose-lowering treatment or pharmacological modulation of histone acetylation rescues DC function and antiviral immunity. Collectively, we highlight a hyperglycaemia-driven metabolic-immune axis orchestrating DC dysfunction during pulmonary viral infection and identify metabolic checkpoints that may be therapeutically exploited in mitigating exacerbated disease in infected diabetics.


Asunto(s)
Células Dendríticas , Complicaciones de la Diabetes , Diabetes Mellitus , Susceptibilidad a Enfermedades , Hiperglucemia , Pulmón , Virosis , Animales , Ratones , Acetilcoenzima A/metabolismo , Acetilación , Cromatina/genética , Cromatina/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Complicaciones de la Diabetes/inmunología , Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Histonas/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/inmunología , Hiperglucemia/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Linfocitos T/inmunología , Virosis/complicaciones , Virosis/inmunología , Virosis/mortalidad , Virus/inmunología , Modelos Animales de Enfermedad , Humanos
3.
Curr Opin Microbiol ; 73: 102287, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36868081

RESUMEN

Caloric restriction (CR) and related time-restricted diets have been popularized as means of preventing metabolic disease while improving general well-being. However, evidence as to their long-term efficacy, adverse effects, and mechanisms of activity remains incompletely understood. The gut microbiota is modulated by such dietary approaches, yet causal evidence to its possible downstream impacts on host metabolism remains elusive. Herein, we discuss the positive and adverse influences of restrictive dietary interventions on gut microbiota composition and function, and their collective impacts on host health and disease risk. We highlight known mechanisms of microbiota influences on the host, such as modulation of bioactive metabolites, while discussing challenges in achieving mechanistic dietary-microbiota insights, including interindividual variability in dietary responses as well as other methodological and conceptual challenges. In all, causally understanding the impact of CR approaches on the gut microbiota may enable to better decode their overall influences on human physiology and disease.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Restricción Calórica , Dieta
4.
Cell Host Microbe ; 31(2): 159-160, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36758512

RESUMEN

In a recent report in Nature, Dohnalová et al., describe a gut-brain axis mechanism in highlighting the importance of the gut microbiota and its derived metabolites in long-term exercise engagement and performance.


Asunto(s)
Encéfalo , Microbioma Gastrointestinal
5.
Clin Microbiol Infect ; 29(6): 682-688, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36191844

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) constitutes a group of auto-inflammatory disorders that impact the gastrointestinal tract and other systemic organs. The gut microbiome contributes to IBD pathology through multiple mechanisms. Bacteriophages (hereafter termed phages) are viruses that are able to specifically infect bacteria. Considered as part of the gut microbiome, phages may impact the bacterial community structure in various clinical contexts. Additionally, exogenous phage administration may represent a means of suppressing IBD-associated pathobionts; however, the utilization of phage therapy remains at an early developmental phase. OBJECTIVES: Herein, we summarize the latest advances in understanding endogenous phage impacts on the gut microbiome in a healthy gut and in IBD. We highlight the prospect of phage utilization as a targeted mode of pathobiont eradication, for preventing and treating IBD manifestations and complications. SOURCES: Selected peer-reviewed publications regarding the role of phages in a healthy gut and in IBD, published between 2013 and 2022. CONTENT: The human gut microbiome is increasingly suggested to play a significant role in the onset and progression of multiple non-communicable diseases such as IBD. Several studies suggest that this effect may be mediated by discrete disease-contributing commensals. However, the eradication of such pathogenic bacteria remains a daunting unmet task. Altered community structure in IBD may be influenced by blooms of phages within the gut bacterial ecosystem. Moreover, combinations of phages specifically targeting disease-contributing pathobiont strain clades may be harnessed as potential eradication treatment preventing and treating IBD, while bearing minimal adverse impacts on the surrounding bacterial microbiome. IMPLICATIONS: Understanding the endogenous phage-gut commensal interactions in a healthy gut and in IBD may enable phage utilization in precision gut microbiome editing, towards treating IBD and other non-communicable microbiome-associated diseases. Nevertheless, developing phage combination-mediated IBD pathobiont eradication treatment modalities will likely necessitate better strain-level bacterial target identification and resolution of treatment-related challenges, such as phage delivery, off-target effects, and bacterial resistance.


Asunto(s)
Bacteriófagos , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Microbiota , Humanos , Enfermedades Inflamatorias del Intestino/terapia , Bacterias
6.
Cell ; 185(16): 2879-2898.e24, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931020

RESUMEN

Human gut commensals are increasingly suggested to impact non-communicable diseases, such as inflammatory bowel diseases (IBD), yet their targeted suppression remains a daunting unmet challenge. In four geographically distinct IBD cohorts (n = 537), we identify a clade of Klebsiella pneumoniae (Kp) strains, featuring a unique antibiotics resistance and mobilome signature, to be strongly associated with disease exacerbation and severity. Transfer of clinical IBD-associated Kp strains into colitis-prone, germ-free, and colonized mice enhances intestinal inflammation. Stepwise generation of a lytic five-phage combination, targeting sensitive and resistant IBD-associated Kp clade members through distinct mechanisms, enables effective Kp suppression in colitis-prone mice, driving an attenuated inflammation and disease severity. Proof-of-concept assessment of Kp-targeting phages in an artificial human gut and in healthy volunteers demonstrates gastric acid-dependent phage resilience, safety, and viability in the lower gut. Collectively, we demonstrate the feasibility of orally administered combination phage therapy in avoiding resistance, while effectively inhibiting non-communicable disease-contributing pathobionts.


Asunto(s)
Bacteriófagos , Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Animales , Colitis/terapia , Humanos , Inflamación/terapia , Enfermedades Inflamatorias del Intestino/terapia , Klebsiella pneumoniae , Ratones
7.
PLoS Comput Biol ; 18(7): e1010212, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35839259

RESUMEN

Longitudinal 'omics analytical methods are extensively used in the evolving field of precision medicine, by enabling 'big data' recording and high-resolution interpretation of complex datasets, driven by individual variations in response to perturbations such as disease pathogenesis, medical treatment or changes in lifestyle. However, inherent technical limitations in biomedical studies often result in the generation of feature-rich and sample-limited datasets. Analyzing such data using conventional modalities often proves to be challenging since the repeated, high-dimensional measurements overload the outlook with inconsequential variations that must be filtered from the data in order to find the true, biologically relevant signal. Tensor methods for the analysis and meaningful representation of multiway data may prove useful to the biological research community by their advertised ability to tackle this challenge. In this study, we present tcam-a new unsupervised tensor factorization method for the analysis of multiway data. Building on top of cutting-edge developments in the field of tensor-tensor algebra, we characterize the unique mathematical properties of our method, namely, 1) preservation of geometric and statistical traits of the data, which enable uncovering information beyond the inter-individual variation that often takes over the focus, especially in human studies. 2) Natural and straightforward out-of-sample extension, making tcam amenable for integration in machine learning workflows. A series of re-analyses of real-world, human experimental datasets showcase these theoretical properties, while providing empirical confirmation of tcam's utility in the analysis of longitudinal 'omics data.


Asunto(s)
Algoritmos , Aprendizaje Automático , Macrodatos , Humanos , Fenotipo , Medicina de Precisión/métodos
9.
Eur J Neurosci ; 56(9): 5413-5427, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35075702

RESUMEN

The incidence of age-related dementia is growing with increased longevity, yet there are currently no disease-modifying therapies for these devastating disorders. Studies over the last several years have led to an evolving awareness of the role of the immune system in supporting brain maintenance and repair, displaying a diverse repertoire of functions while orchestrating the crosstalk between the periphery and the brain. Here, we provide insights into the current understanding of therapeutic targets that could be adopted to modulate immune cell fate, either systemically or locally, to defeat brain aging and neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Humanos , Longevidad , Sistema Inmunológico
10.
Nature ; 600(7890): 713-719, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34880502

RESUMEN

Cigarette smoking constitutes a leading global cause of morbidity and preventable death1, and most active smokers report a desire or recent attempt to quit2. Smoking-cessation-induced weight gain (SCWG; 4.5 kg reported to be gained on average per 6-12 months, >10 kg year-1 in 13% of those who stopped smoking3) constitutes a major obstacle to smoking abstinence4, even under stable5,6 or restricted7 caloric intake. Here we use a mouse model to demonstrate that smoking and cessation induce a dysbiotic state that is driven by an intestinal influx of cigarette-smoke-related metabolites. Microbiome depletion induced by treatment with antibiotics prevents SCWG. Conversely, fecal microbiome transplantation from mice previously exposed to cigarette smoke into germ-free mice naive to smoke exposure induces excessive weight gain across diets and mouse strains. Metabolically, microbiome-induced SCWG involves a concerted host and microbiome shunting of dietary choline to dimethylglycine driving increased gut energy harvest, coupled with the depletion of a cross-regulated weight-lowering metabolite, N-acetylglycine, and possibly by the effects of other differentially abundant cigarette-smoke-related metabolites. Dimethylglycine and N-acetylglycine may also modulate weight and associated adipose-tissue immunity under non-smoking conditions. Preliminary observations in a small cross-sectional human cohort support these findings, which calls for larger human trials to establish the relevance of this mechanism in active smokers. Collectively, we uncover a microbiome-dependent orchestration of SCWG that may be exploitable to improve smoking-cessation success and to correct metabolic perturbations even in non-smoking settings.


Asunto(s)
Microbioma Gastrointestinal , Cese del Hábito de Fumar , Aumento de Peso , Animales , Estudios Transversales , Disbiosis/etiología , Disbiosis/metabolismo , Disbiosis/patología , Ratones , Modelos Animales , Fumar/metabolismo , Fumar/patología
11.
Gut Microbes ; 11(6): 1833-1841, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32501768

RESUMEN

The intestinal microbiota may be involved, through metabolic gut-brain interactions, in a variety of neurological conditions. In this addendum, we summarize the findings of our recent study investigating the potentially modulatory influence of the microbiome in a transgenic ALS mouse model, and the possible application to human disease. We found that transgenic mice show evidence of dysbiosis, even at the pre-symptomatic stage, and have a more severe disease course under germ-free conditions or after receiving broad-spectrum antibiotics. We demonstrated that Akkermansia muciniphila ameliorated the disease in mice and that this may be due to the production of nicotinamide. We then conducted a preliminary study in human ALS and identified functionally similar alterations within the metagenome. Furthermore, we found that patients with ALS had lower systemic and CSF levels of nicotinamide, suggesting that the changes observed in the mouse model may be relevant to human disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/microbiología , Microbioma Gastrointestinal , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Modelos Animales de Enfermedad , Disbiosis/microbiología , Humanos
12.
Nature ; 572(7770): 474-480, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31330533

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disorder, in which the clinical manifestations may be influenced by genetic and unknown environmental factors. Here we show that ALS-prone Sod1 transgenic (Sod1-Tg) mice have a pre-symptomatic, vivarium-dependent dysbiosis and altered metabolite configuration, coupled with an exacerbated disease under germ-free conditions or after treatment with broad-spectrum antibiotics. We correlate eleven distinct commensal bacteria at our vivarium with the severity of ALS in mice, and by their individual supplementation into antibiotic-treated Sod1-Tg mice we demonstrate that Akkermansia muciniphila (AM) ameliorates whereas Ruminococcus torques and Parabacteroides distasonis exacerbate the symptoms of ALS. Furthermore, Sod1-Tg mice that are administered AM are found to accumulate AM-associated nicotinamide in the central nervous system, and systemic supplementation of nicotinamide improves motor symptoms and gene expression patterns in the spinal cord of Sod1-Tg mice. In humans, we identify distinct microbiome and metabolite configurations-including reduced levels of nicotinamide systemically and in the cerebrospinal fluid-in a small preliminary study that compares patients with ALS with household controls. We suggest that environmentally driven microbiome-brain interactions may modulate ALS in mice, and we call for similar investigations in the human form of the disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/microbiología , Esclerosis Amiotrófica Lateral/fisiopatología , Microbioma Gastrointestinal/fisiología , Niacinamida/metabolismo , Akkermansia , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Antibacterianos/farmacología , Modelos Animales de Enfermedad , Disbiosis , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Vida Libre de Gérmenes , Humanos , Longevidad , Masculino , Ratones , Ratones Transgénicos , Niacinamida/biosíntesis , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Tasa de Supervivencia , Simbiosis/efectos de los fármacos , Verrucomicrobia/metabolismo , Verrucomicrobia/fisiología
13.
Cell Rep ; 26(13): 3586-3599.e7, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917314

RESUMEN

The tuberculosis (TB) bacillus, Mycobacterium tuberculosis (Mtb), and HIV-1 act synergistically; however, the mechanisms by which Mtb exacerbates HIV-1 pathogenesis are not well known. Using in vitro and ex vivo cell culture systems, we show that human M(IL-10) anti-inflammatory macrophages, present in TB-associated microenvironment, produce high levels of HIV-1. In vivo, M(IL-10) macrophages are expanded in lungs of co-infected non-human primates, which correlates with disease severity. Furthermore, HIV-1/Mtb co-infected patients display an accumulation of M(IL-10) macrophage markers (soluble CD163 and MerTK). These M(IL-10) macrophages form direct cell-to-cell bridges, which we identified as tunneling nanotubes (TNTs) involved in viral transfer. TNT formation requires the IL-10/STAT3 signaling pathway, and targeted inhibition of TNTs substantially reduces the enhancement of HIV-1 cell-to-cell transfer and overproduction in M(IL-10) macrophages. Our study reveals that TNTs facilitate viral transfer and amplification, thereby promoting TNT formation as a mechanism to be explored in TB/AIDS potential therapeutics.


Asunto(s)
Infecciones por VIH/complicaciones , Interleucina-10/metabolismo , Macrófagos/patología , Nanotubos , Factor de Transcripción STAT3/metabolismo , Tuberculosis Pulmonar/complicaciones , Adulto , Anciano , Animales , Células Cultivadas , Coinfección/patología , Coinfección/virología , Femenino , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Infecciones por VIH/virología , Humanos , Macaca mulatta , Activación de Macrófagos , Macrófagos/virología , Masculino , Persona de Mediana Edad , Mycobacterium tuberculosis , Transducción de Señal , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Replicación Viral , Adulto Joven
15.
J Mol Cell Cardiol ; 125: 149-161, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30393107

RESUMEN

Coxsackievirus B3 (CVB3) is a globally prevalent enterovirus of the Picornaviridae family that is frequently associated with viral myocarditis (VM). Neutrophils, as first responders, may be key cells in determining viral disease outcomes; however, neutrophils have been poorly studied with respect to viral infection. Although neutrophils have been ascribed a relevant role in early cardiac inflammation, their precise role in CVB3 infection has not yet been evaluated. In this study, we aimed to determine if the interaction between human neutrophils and CVB3 could lead to viral replication and/or modulation of neutrophil survival and biological functions, and whether neutrophil depletion in a murine model has a beneficial or harmful effect on CVB3 infection. Our results show that CVB3 interacted with but did not replicate in human neutrophils. Neutrophils recognized CVB3 mainly through endosomal TLR-8, and infection triggered NFκB activation. Virus internalization resulted in increased cell survival, up-regulation of CD11b, enhanced adhesion to fibrinogen and fibronectin, and the secretion of IL-6, IL-1ß, TNF-α, and IL-8. Supernatants from infected neutrophils exerted chemotactic activity partly mediated by IL-8. The infected neutrophils released myeloperoxidase and triggered neutrophil extracellular trap formation in the presence of TNF-α. In mice infected with CVB3, viral RNA was detected in neutrophils as well as in mononuclear cells. After neutrophil depletion, mice showed reduced VM reflected by a reduction in viral titers, cell exudates, and CCL-2 mRNA levels, as well as the abrogation of reactive cardiomyocyte hypertrophy. Our results indicate that neutrophils have relevant direct and indirect roles in the pathogenesis of CVB3-induced VM.


Asunto(s)
Infecciones por Coxsackievirus/metabolismo , Miocarditis/metabolismo , Miocarditis/virología , Neutrófilos/metabolismo , Animales , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/virología , Enterovirus Humano B/inmunología , Enterovirus Humano B/patogenicidad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-1beta/metabolismo , Miocarditis/inmunología , Miocardio/inmunología , Miocardio/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/metabolismo , Replicación Viral/fisiología
16.
Front Immunol ; 9: 459, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593722

RESUMEN

The ability of Mycobacterium tuberculosis (Mtb) to persist in its human host relies on numerous immune evasion strategies, such as the deregulation of the lipid metabolism leading to the formation of foamy macrophages (FM). Yet, the specific host factors leading to the foamy phenotype of Mtb-infected macrophages remain unknown. Herein, we aimed to address whether host cytokines contribute to FM formation in the context of Mtb infection. Our approach is based on the use of an acellular fraction of tuberculous pleural effusions (TB-PE) as a physiological source of local factors released during Mtb infection. We found that TB-PE induced FM differentiation as observed by the increase in lipid bodies, intracellular cholesterol, and expression of the scavenger receptor CD36, as well as the enzyme acyl CoA:cholesterol acyl transferase (ACAT). Importantly, interleukin-10 (IL-10) depletion from TB-PE prevented the augmentation of all these parameters. Moreover, we observed a positive correlation between the levels of IL-10 and the number of lipid-laden CD14+ cells among the pleural cells in TB patients, demonstrating that FM differentiation occurs within the pleural environment. Downstream of IL-10 signaling, we noticed that the transcription factor signal transducer and activator of transcription 3 was activated by TB-PE, and its chemical inhibition prevented the accumulation of lipid bodies and ACAT expression in macrophages. In terms of the host immune response, TB-PE-treated macrophages displayed immunosuppressive properties and bore higher bacillary loads. Finally, we confirmed our results using bone marrow-derived macrophage from IL-10-/- mice demonstrating that IL-10 deficiency partially prevented foamy phenotype induction after Mtb lipids exposure. In conclusion, our results evidence a role of IL-10 in promoting the differentiation of FM in the context of Mtb infection, contributing to our understanding of how alterations of the host metabolic factors may favor pathogen persistence.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/inmunología , Regulación Enzimológica de la Expresión Génica/inmunología , Interleucina-10/inmunología , Mycobacterium tuberculosis/inmunología , Derrame Pleural/inmunología , Factor de Transcripción STAT3/inmunología , Esterol O-Aciltransferasa , Tuberculosis Pleural/inmunología , Regulación hacia Arriba/inmunología , Acetil-CoA C-Acetiltransferasa/genética , Animales , Femenino , Células Espumosas , Humanos , Interleucina-10/genética , Masculino , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/genética , Derrame Pleural/genética , Derrame Pleural/patología , Factor de Transcripción STAT3/genética , Tuberculosis Pleural/genética , Tuberculosis Pleural/patología
17.
Mediators Inflamm ; 2017: 2810606, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28852268

RESUMEN

M strain, the most prevalent multidrug-resistant strain of Mycobacterium tuberculosis (Mtb) in Argentina, has mounted mechanisms to evade innate immune response. The role of human bronchial epithelium in Mtb infection remains unknown as well as its crosstalk with neutrophils (PMN). In this work, we evaluate whether M and H37Rv strains invade and replicate within bronchial epithelial cell line Calu-6 and how conditioned media (CM) derived from infected cells alter PMN responses. We demonstrated that M infects and survives within Calu-6 without promoting death. CM from M-infected Calu-6 (M-CM) did not attract PMN in correlation with its low IL-8 content compared to H37Rv-CM. Also, PMN activation and ROS production in response to irradiated H37Rv were impaired after treatment with M-CM due to the lack of TNF-α. Interestingly, M-CM increased H37Rv replication in PMN which would allow the spreading of mycobacteria upon PMN death and sustain IL-8 release. Thus, our results indicate that even at low invasion/replication rate within Calu-6, M induces the secretion of factors altering the crosstalk between these nonphagocytic cells and PMN, representing an evasion mechanism developed by M strain to persist in the host. These data provide new insights on the role of bronchial epithelium upon M infection.


Asunto(s)
Interleucina-8/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Quimiocinas/metabolismo , Quimiotaxis/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Citometría de Flujo , Humanos , Inmunidad Innata/efectos de los fármacos , Fagocitosis/efectos de los fármacos
18.
Int J Med Microbiol ; 306(7): 541-553, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27394957

RESUMEN

Tuberculosis (TB) is a complex disease, and the success of the bacterium depends on its ability to evade the immune response. Previously, we determined that Mycobacterium tuberculosis (Mtb) impairs the function of dendritic cells (DC), promoting the generation of cells that are poor stimulators of mycobacterial antigen-specific CD4T cells, which are required to control this persistent infection. In this study, we aimed to determine the mechanisms by which monocyte-derived DCs differentiated in the presence of Mtb (MtbDC) may impact on the proliferation of specific anti-mycobacterial T cells. We found that the presence of Mtb during monocyte-derived DC differentiation favours T helper (Th) 2 and Th17 polarization, in detriment of a Th1 response, compared to DC mature with Mtb. The bias on T cell polarization was associated to the profile of C-type lectin receptors expression found in MtbDC (DC-SIGNlow/MRlow/Dectin-1high). Alternatively, MtbDC release Mtb antigens (Ag) that can be taken up and presented by bystander DC, promoting the proliferation of CD4T cells, but to a lesser extent than direct presentation by Mtb-matured DC. In summary, we have further characterized the generation of MtbDC as an effective evasion strategy driven by the pathogen, leading to the inhibition of Ag-presentation and bias of T cell polarization towards Th2 and Th17 profiles, features which partially explain the persistence of Mtb in the host.


Asunto(s)
Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Mycobacterium tuberculosis/inmunología , Células Th17/inmunología , Células Th2/inmunología , Adulto , Donantes de Sangre , Proliferación Celular , Voluntarios Sanos , Humanos , Persona de Mediana Edad
19.
Clin Sci (Lond) ; 129(4): 319-30, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25858460

RESUMEN

Circulating monocytes (Mo) play an essential role in the host immune response to chronic infections. We previously demonstrated that CD16(pos) Mo were expanded in TB (tuberculosis) patients, correlated with disease severity and were refractory to dendritic cell differentiation. In the present study, we investigated whether human Mo subsets (CD16(neg) and CD16(pos)) differed in their ability to influence the early inflammatory response against Mycobacterium tuberculosis. We first evaluated the capacity of the Mo subsets to migrate and engage a microbicidal response in vitro. Accordingly, CD16(neg) Mo were more prone to migrate in response to different mycobacteria-derived gradients, were more resistant to M. tuberculosis intracellular growth and produced higher reactive oxygen species than their CD16(pos) counterpart. To assess further the functional dichotomy among the human Mo subsets, we carried out an in vivo analysis by adapting a hybrid mouse model (SCID/Beige, where SCID is severe combined immunodeficient) to transfer each Mo subset, track their migratory fate during M. tuberculosis infection, and determine their impact on the host immune response. In M. tuberculosis-infected mice, the adoptively transferred CD16(neg) Mo displayed a higher lung migration index, induced a stronger pulmonary infiltration of murine leucocytes expressing pro- and anti-inflammatory cytokines, and significantly decreased the bacterial burden, in comparison with CD16(pos) Mo. Collectively, our results indicate that human Mo subsets display divergent biological roles in the context of M. tuberculosis infection, a scenario in which CD16(neg) Mo may contribute to the anti-mycobacterial immune response, whereas CD16(pos) Mo might promote microbial resilience, shedding light on a key aspect of the physiopathology of TB disease.


Asunto(s)
Pulmón/inmunología , Monocitos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Carga Bacteriana , Células Cultivadas , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Interacciones Huésped-Patógeno , Humanos , Pulmón/metabolismo , Pulmón/microbiología , Ratones SCID , Monocitos/clasificación , Monocitos/metabolismo , Monocitos/microbiología , Monocitos/trasplante , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Estallido Respiratorio , Factores de Tiempo , Tuberculosis Pulmonar/sangre , Tuberculosis Pulmonar/microbiología
20.
PLoS One ; 9(5): e97837, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24836916

RESUMEN

In human tuberculosis (TB), CD8+ T cells contribute to host defense by the release of Th1 cytokines and the direct killing of Mycobacterium tuberculosis (Mtb)-infected macrophages via granule exocytosis pathway or the engagement of receptors on target cells. Previously we demonstrated that strain M, the most prevalent multidrug-resistant (MDR) Mtb strain in Argentine, is a weak inducer of IFN-γ and elicits a remarkably low CD8-dependent cytotoxic T cell activity (CTL). In contrast, the closely related strain 410, which caused a unique case of MDR-TB, elicits a CTL response similar to H37Rv. In this work we extend our previous study investigating some parameters that can account for this discrepancy. We evaluated the expressions of the lytic molecules perforin, granzyme B and granulysin and the chemokine CCL5 in CD8+ T cells as well as activation markers CD69 and CD25 and IL-2 expression in CD4+ and CD8+ T cells stimulated with strains H37Rv, M and 410. Our results demonstrate that M-stimulated CD8+ T cells from purified protein derivative positive healthy donors show low intracellular expression of perforin, granzyme B, granulysin and CCL5 together with an impaired ability to form conjugates with autologous M-pulsed macrophages. Besides, M induces low CD69 and IL-2 expression in CD4+ and CD8+ T cells, being CD69 and IL-2 expression closely associated. Furthermore, IL-2 addition enhanced perforin and granulysin expression as well as the degranulation marker CD107 in M-stimulated CD8+ T cells, making no differences with cells stimulated with strains H37Rv or 410. Thus, our results highlight the role of IL-2 in M-induced CTL activity that drives the proper activation of CD8+ T cells as well as CD4+ T cells collaboration.


Asunto(s)
Citotoxicidad Inmunológica , Farmacorresistencia Bacteriana Múltiple , Activación de Linfocitos , Mycobacterium tuberculosis/inmunología , Linfocitos T Citotóxicos/inmunología , Adulto , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Femenino , Granzimas/genética , Granzimas/metabolismo , Humanos , Interleucina-2/genética , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Masculino , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/genética , Perforina/genética , Perforina/metabolismo , Linfocitos T Citotóxicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA