Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 16(10): e1008929, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33002063

RESUMEN

The ability of Mycobacterium tuberculosis (Mtb) to persist inside host cells relies on metabolic adaptation, like the accumulation of lipid bodies (LBs) in the so-called foamy macrophages (FM), which are favorable to Mtb. The activation state of macrophages is tightly associated to different metabolic pathways, such as lipid metabolism, but whether differentiation towards FM differs between the macrophage activation profiles remains unclear. Here, we aimed to elucidate whether distinct macrophage activation states exposed to a tuberculosis-associated microenvironment or directly infected with Mtb can form FM. We showed that the triggering of signal transducer and activator of transcription 6 (STAT6) in interleukin (IL)-4-activated human macrophages (M(IL-4)) prevents FM formation induced by pleural effusion from patients with tuberculosis. In these cells, LBs are disrupted by lipolysis, and the released fatty acids enter the ß-oxidation (FAO) pathway fueling the generation of ATP in mitochondria. Accordingly, murine alveolar macrophages, which exhibit a predominant FAO metabolism, are less prone to become FM than bone marrow derived-macrophages. Interestingly, direct infection of M(IL-4) macrophages with Mtb results in the establishment of aerobic glycolytic pathway and FM formation, which could be prevented by FAO activation or inhibition of the hypoxia-inducible factor 1-alpha (HIF-1α)-induced glycolytic pathway. In conclusion, our results demonstrate that Mtb has a remarkable capacity to induce FM formation through the rewiring of metabolic pathways in human macrophages, including the STAT6-driven alternatively activated program. This study provides key insights into macrophage metabolism and pathogen subversion strategies.


Asunto(s)
Células Espumosas/microbiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metabolismo de los Lípidos , Macrófagos/microbiología , Mycobacterium tuberculosis/patogenicidad , Animales , Gotas Lipídicas/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/fisiología , Tuberculosis/microbiología
2.
PLoS Pathog ; 13(8): e1006527, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28767704

RESUMEN

Despite eliciting a potent CD8+ T cell response, Brucella abortus is able to persist and establish a chronic infection inside its host. We have previously reported that the infection of human monocytes/macrophages with B. abortus inhibits the IFN-γ-induced MHC-I cell surface expression down-modulating cytotoxic CD8+ T cell responses. MHC-I down-modulation depends on bacterial viability and results from the capacity of B. abortus to retain the MHC-I molecules within the Golgi apparatus. Furthermore, we recently demonstrated that epidermal growth factor receptor (EGFR) pathway is involved in this phenomenon and that this is an early event during infection. However, the components and mechanisms whereby B. abortus is able to down-modulate MHC-I remained to be elucidated. In this study we demonstrated that the down-modulation of MHC-I expression is not mediated by well-known Brucella virulence factors but instead by B. abortus RNA, a PAMP associated to viability (vita-PAMP). Surprisingly, completely degraded RNA was also able to inhibit MHC-I expression to the same extent as intact RNA. Accordingly, B. abortus RNA and its degradation products were able to mimic the MHC-I intracellular retention within the Golgi apparatus observed upon infection. We further demonstrated that TLR8, a single-stranded RNA and RNA degradation products sensor, was involved in MHC-I inhibition. On the other hand, neutralization of the EGFR reversed the MHC-I inhibition, suggesting a connection between the TLR8 and EGFR pathways. Finally, B. abortus RNA-treated macrophages display diminished capacity of antigen presentation to CD8+ T cells. Overall, our results indicate that the vita-PAMP RNA as well as its degradation products constitute novel virulence factors whereby B. abortus, by a TLR8-dependent mechanism and through the EGFR pathway, inhibits the IFN-γ-induced MHC-I surface expression on human monocytes/macrophages. Thus, bacteria can hide within infected cells and avoid the immunological surveillance of cytotoxic CD8+ T cells.


Asunto(s)
Brucelosis/inmunología , Receptores ErbB/inmunología , Evasión Inmune/inmunología , Monocitos/inmunología , ARN Bacteriano/inmunología , Receptor Toll-Like 8/inmunología , Animales , Brucella abortus/inmunología , Reactividad Cruzada/inmunología , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Monocitos/microbiología , Transducción de Señal/inmunología
3.
Immunology ; 151(1): 122-135, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28106253

RESUMEN

Beside its key diagnostic value, the humoral immune response is thought to play a protective role in hantavirus pulmonary syndrome. However, little is known about the cell source of these antibodies during ongoing human infection. Herein we characterized B-cell subsets circulating in Andes-virus-infected patients. A notable potent plasmablast (PB) response that increased 100-fold over the baseline levels was observed around 1 week after the onset of symptoms. These PB present a CD3neg CD19low CD20neg CD38hi CD27hi CD138+/- IgA+/- surface phenotype together with the presence of cytoplasmic functional immunoglobulins. They are large lymphocytes (lymphoblasts) morphologically coincident with the 'immunoblast-like' cells that have been previously described during blood cytology examinations of hantavirus-infected patients. Immunoreactivity analysis of white blood cell lysates suggests that some circulating PB are virus-specific but we also observed a significant increase of reactivity against virus-unrelated antigens, which suggests a possible bystander effect by polyclonal B-cell activation. The presence of this large and transient PB response raises the question as to whether these cells might have a protective or pathological role during the ongoing hantavirus pulmonary syndrome and suggest their practical application as a diagnostic/prognostic biomarker.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Síndrome Pulmonar por Hantavirus/inmunología , Orthohantavirus/inmunología , Células Plasmáticas/inmunología , Células Precursoras de Linfocitos B/inmunología , Enfermedad Aguda , Adulto , Anticuerpos Antivirales/sangre , Antígenos CD/metabolismo , Autoantígenos/inmunología , Subgrupos de Linfocitos B/virología , Biomarcadores/metabolismo , Proliferación Celular , Femenino , Síndrome Pulmonar por Hantavirus/diagnóstico , Humanos , Inmunoglobulina A/metabolismo , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Células Plasmáticas/virología , Células Precursoras de Linfocitos B/virología , Adulto Joven
4.
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
5.
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
6.
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
8.
Eur J Immunol ; 43(2): 335-47, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23192690

RESUMEN

Tuberculosis (TB) is one of the world's most pernicious diseases mainly due to immune evasion strategies displayed by its causative agent Mycobacterium tuberculosis (Mtb). Blood monocytes (Mos) represent an important source of DCs during chronic infections; consequently, the alteration of their differentiation constitutes an escape mechanism leading to mycobacterial persistence. We evaluated whether the CD16(+)/CD16(-) Mo ratio could be associated with the impaired Mo differentiation into DCs found in TB patients. The phenotype and ability to stimulate Mtb-specific memory clones DCs from isolated Mo subsets were assessed. We found that CD16(-) Mos differentiated into CD1a(+) DC-SIGN(high) cells achieving an efficient recall response, while CD16(+) Mos differentiated into a CD1a(-) DC-SIGN(low) population characterized by a poor mycobacterial Ag-presenting capacity. The high and sustained phosphorylated p38 expression observed in CD16(+) Mos was involved in the altered DC profile given that its blockage restored DC phenotype and its activation impaired CD16(-) Mo differentiation. Furthermore, depletion of CD16(+) Mos indeed improved the differentiation of Mos from TB patients toward CD1a(+) DC-SIGN(high) DCs. Therefore, Mos from TB patients are less prone to differentiate into DCs due to their increased proportion of CD16(+) Mos, suggesting that during Mtb infection Mo subsets may have different fates after entering the lungs.


Asunto(s)
Células Dendríticas/patología , Monocitos/patología , Receptores de IgG/metabolismo , Tuberculosis/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adulto , Antígenos CD1/inmunología , Antígenos CD1/metabolismo , Diferenciación Celular/inmunología , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Monocitos/enzimología , Monocitos/inmunología , Monocitos/metabolismo , Mycobacterium tuberculosis/inmunología , Receptores de IgG/inmunología , Transducción de Señal/inmunología , Transducción de Señal/fisiología , Tuberculosis/enzimología , Tuberculosis/metabolismo , Tuberculosis/microbiología , Adulto Joven , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
9.
Clin Sci (Lond) ; 127(6): 391-403, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24689690

RESUMEN

DTH (delayed type hypersensitivity) reactions are secondary cellular immune responses that appear 24-72 h after antigen exposure. Tuberculous pleurisy is a common manifestation of extrapulmonary TB (tuberculosis) and is considered a human model of Th1-mediated DTH. In order to identify functional cross-talk among cellular populations sited in this inflammatory microenvironment, we analysed phenotypic and functional features of human B-cells isolated from the PF (pleural fluid) of TB patients. Freshly isolated PF-B-cells displayed a lower expression of CD20, CD1d and HLA-DR, and a higher expression of CD95, CD38, CD25, CXCR3 (CXC chemokine receptor 3) and CXCR4 (CXC chemokine receptor 4) than their PB (peripheral blood) counterparts, suggesting a non-classical in situ activation. Although memory PF-T-cell frequencies were increased, the frequencies of memory PF-B-cells were not. We demonstrated that, upon stimulation with γ-irradiated M. tuberculosis, mycobacterially secreted proteins or a lectin mitogen, PF-B-cells had a strong activation and produced IL-10 by a mechanism that was dependent on bystander activation of CD19(-) PF cells. Besides, within PF cells, B-cells diminished in vitro M. tuberculosis-induced IFN (interferon)-γ production by T-cells and NK (natural killer) cells in an IL-10-dependent manner. Finally, we found that the lower the frequency of B-cells, the higher the ratio of IFN-γ/IL-10 within PF. Thus our results suggest that B-cells can regulate a human DTH reaction induced by M. tuberculosis.


Asunto(s)
Linfocitos B/metabolismo , Hipersensibilidad Tardía/inmunología , Interferón gamma/biosíntesis , Células Asesinas Naturales/metabolismo , Linfocitos T/metabolismo , Tuberculosis Pleural/microbiología , Linfocitos B/inmunología , Humanos , Inmunidad Celular/inmunología , Interleucina-10/inmunología , Células Asesinas Naturales/inmunología , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Tuberculosis Pleural/inmunología
10.
J Immunol ; 189(6): 3121-9, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22869905

RESUMEN

The role of macrophage-inducible C-type lectin Mincle in lung innate immunity against mycobacterial infection is incompletely defined. In this study, we show that wild-type (WT) mice responded with a delayed Mincle induction on resident alveolar macrophages and newly immigrating exudate macrophages to infection with Mycobacterium bovis bacillus Calmette-Guérin (BCG), peaking by days 14-21 posttreatment. As compared with WT mice, Mincle knockout (KO) mice exhibited decreased proinflammatory mediator responses and leukocyte recruitment upon M. bovis BCG challenge, and they demonstrated increased mycobacterial loads in pulmonary and extrapulmonary organ systems. Secondary mycobacterial infection on day 14 after primary BCG challenge led to increased cytokine gene expression in sorted alveolar macrophages of WT mice, but not Mincle KO mice, resulting in substantially reduced alveolar neutrophil recruitment and increased mycobacterial loads in the lungs of Mincle KO mice. Collectively, these data show that WT mice respond with a relatively late Mincle expression on lung sentinel cells to M. bovis BCG infection. Moreover, M. bovis BCG-induced upregulation of C-type lectin Mincle on professional phagocytes critically shapes antimycobacterial responses in both pulmonary and extrapulmonary organ systems of mice, which may be important for elucidating the role of Mincle in the control of mycobacterial dissemination in mice.


Asunto(s)
Inmunidad Innata , Lectinas Tipo C/fisiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Proteínas de la Membrana/fisiología , Mycobacterium bovis/inmunología , Tuberculosis/inmunología , Tuberculosis/metabolismo , Animales , Inmunidad Innata/genética , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Lectinas Tipo C/biosíntesis , Lectinas Tipo C/deficiencia , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/microbiología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/microbiología , Neutrófilos/patología , Transducción de Señal/genética , Transducción de Señal/inmunología , Tuberculosis/prevención & control
11.
Elife ; 122024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38922679

RESUMEN

During tuberculosis (TB), migration of dendritic cells (DCs) from the site of infection to the draining lymph nodes is known to be impaired, hindering the rapid development of protective T-cell-mediated immunity. However, the mechanisms involved in the delayed migration of DCs during TB are still poorly defined. Here, we found that infection of DCs with Mycobacterium tuberculosis (Mtb) triggers HIF1A-mediated aerobic glycolysis in a TLR2-dependent manner, and that this metabolic profile is essential for DC migration. In particular, the lactate dehydrogenase inhibitor oxamate and the HIF1A inhibitor PX-478 abrogated Mtb-induced DC migration in vitro to the lymphoid tissue-specific chemokine CCL21, and in vivo to lymph nodes in mice. Strikingly, we found that although monocytes from TB patients are inherently biased toward glycolysis metabolism, they differentiate into poorly glycolytic and poorly migratory DCs compared with healthy subjects. Taken together, these data suggest that because of their preexisting glycolytic state, circulating monocytes from TB patients are refractory to differentiation into migratory DCs, which may explain the delayed migration of these cells during the disease and opens avenues for host-directed therapies for TB.


Asunto(s)
Movimiento Celular , Células Dendríticas , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia , Monocitos , Mycobacterium tuberculosis , Tuberculosis , Células Dendríticas/metabolismo , Células Dendríticas/inmunología , Monocitos/metabolismo , Monocitos/inmunología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mycobacterium tuberculosis/inmunología , Animales , Tuberculosis/inmunología , Tuberculosis/metabolismo , Tuberculosis/microbiología , Ratones , Receptor Toll-Like 2/metabolismo , Ratones Endogámicos C57BL , Femenino
12.
Clin Dev Immunol ; 2012: 152546, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22778761

RESUMEN

Tuberculosis pathogenesis was earlier thought to be mainly related to the host but now it appears to be clear that bacterial factors are also involved. Genetic variability of Mycobacterium tuberculosis (Mtb) could be slight but it may lead to sharp phenotypic differences. We have previously reported that nonopsonized Mtb H37Rv induce apoptosis of polymorphonuclear neutrophils (PMNs) by a mechanism that involves the p38 pathway. Here we evaluated the capability to induce PMN apoptosis of two prevalent Mtb lineages in Argentina, the Latin America and Mediterranean (LAM), and Haarlem, using the H37Rv as a reference strain. Results showed that LAM strains strongly induced apoptosis of PMN which correlated with the induction of reactive oxygen species (ROS) production and p38 activation. Interestingly, the highly prosperous multidrug-resistant M strain, belonging to the Haarlem lineage, lacked the ability to activate and to induce PMN apoptosis as a consequence of (1) a weak ROS production and (2) the contribution of antiapoptotic mechanisms mediated at least by ERK. Although with less skill, M is able to enter the PMN so that phenotypic differences could lead PMN to be a reservoir allowing some pathogens to prevail and persist over other strains in the community.


Asunto(s)
Apoptosis/inmunología , Mycobacterium tuberculosis/inmunología , Neutrófilos/inmunología , Estallido Respiratorio/inmunología , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Moléculas de Adhesión Celular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Ligadas a GPI/metabolismo , Humanos , Mycobacterium tuberculosis/aislamiento & purificación , Neutrófilos/metabolismo , Neutrófilos/microbiología , Fagocitosis/inmunología , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Infect Dis ; 204(7): 1054-64, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21881121

RESUMEN

BACKGROUND: The proinflammatory cytokine interleukin 17 (IL-17) plays an important role in immune responses but it is also associated with tissue-damaging inflammation. So, we evaluated the ability of Mycobacterium tuberculosis clinical isolates to induce IL-17 in tuberculosis (TB) patients and in healthy human tuberculin reactors (PPD(+)HD). METHODS: IL-17, interferon γ (IFN-γ), and interleukin 23 (IL-23) receptor expression were evaluated ex vivo and cultured peripheral blood mononuclear cells from TB and PPD(+)HD stimulated with irradiated clinical isolates from multidrug resistant (MDR) outbreaks M (Haarlem family) and Ra (Latin American-Mediterranean family), as well as drug-susceptible isolates belonging to the same families and laboratory strain H37Rv for 48 hours in T-cell subsets by flow cytometry. RESULTS: We observed that: (1) MDR strains M and Ra are stronger IL-17 inducers than drug-susceptible Mtb strains of the Haarlem and Latin American-Mediterranean families, (2) MDR-TB patients show the highest IL-17 expression that is independent on the strain, (3) IL-17 expression is dependent on CD4(+) and CD8(+) T cells associates with persistently high antigen load. CONCLUSIONS: IL-17--producing T cells could play an immunopathological role in MDR-TB promoting severe tissue damage, which may be associated with the low effectiveness of the second-line drugs employed in the treatment.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Mycobacterium tuberculosis/inmunología , Subgrupos de Linfocitos T/inmunología , Tuberculosis Resistente a Múltiples Medicamentos/inmunología , Adulto , Células Cultivadas , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Masculino , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Interleucina/metabolismo , Subgrupos de Linfocitos T/metabolismo , Adulto Joven
14.
J Leukoc Biol ; 112(1): 173-184, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34811790

RESUMEN

We previously demonstrated that Bordetella pertussis, the etiologic agent of whooping cough, is able to survive inside human macrophages. The aim of this study was to examine the influence of macrophage polarization in the development of B. pertussis intracellular infections. To this end, primary human monocytes were differentiated into M1, M2a, or M2c macrophages and further infected with B. pertussis. Infected M1 macrophages showed a proinflammatory response evidenced by the production of TNF-α, IL-12p70, and IL-6. Conversely, infection of M2a and M2c macrophages did not induce TNF-α, IL-12p70, nor IL-6 at any time postinfection but showed a significant increase of M2 markers, such as CD206, CD163, and CD209. Interestingly, anti-inflammatory cytokines, like IL-10 and TGF-ß, were induced after infection in the 3 macrophage phenotypes. B. pertussis phagocytosis by M1 macrophages was lower than by M2 phenotypes, which may be ascribed to differences in the expression level of B. pertussis docking molecules on the surface of the different phenotypes. Intracellular bactericidal activity was found to be significantly higher in M1 than in M2a or M2c cells, but live bacteria were still detected within the 3 phenotypes at the late time points after infection. In summary, this study shows that intracellular B. pertussis is able to survive regardless of the macrophage activation program, but its intracellular survival proved higher in M2 compared with the M1 macrophages, being M2c the best candidate to develop into a niche of persistence for B. pertussis.


Asunto(s)
Activación de Macrófagos , Tos Ferina , Bordetella pertussis , Humanos , Interleucina-6/metabolismo , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Tos Ferina/metabolismo
15.
PLoS Negl Trop Dis ; 16(11): e0010950, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36441810

RESUMEN

Monocytes and macrophages play a central role in chronic brucellosis. Brucella abortus (Ba) is an intracellular pathogen that survives inside these cells. On the other hand, macrophages could be differentiated into classical (M1), alternative (M2) or other less-identified profiles. We have previously shown that Ba RNA (a bacterial viability-associated PAMP or vita-PAMP) is a key molecule by which Ba can evade the host immune response. However, we did not know if macrophages could be polarized by this vita-PAMP. To assess this, we used two different approaches: we evaluated if Ba RNA per se was able to differentiate macrophages to M1 or M2 or, given that Ba survives inside macrophages once a Th1 response is established (i.e., in the presence of IFN-γ), we also analysed if Ba RNA could interfere with M1 polarization. We found that Ba RNA alone does not polarize to M1 or M2 but activates human macrophages instead. However, our results show that Ba RNA does interfere with M1 polarization while they are being differentiated. This vita-PAMP diminished the M1-induced CD64, and MHC-II surface expression on macrophages at 48 h. This phenomenon was not associated with an alternative activation of these cells (M2), as shown by unchanged CD206, DC-SIGN and CD163 surface expression. When evaluating glucose metabolism, we found that Ba RNA did not modify M1 glucose consumption or lactate production. However, production of Nitrogen Reactive Species (NRS) did diminish in Ba RNA-treated M1 macrophages. Overall, our results show that Ba RNA could alter the proper immune response set to counterattack the bacteria that could persist in the host establishing a chronic infection.


Asunto(s)
Brucella abortus , ARN , Humanos , Brucella abortus/genética
16.
J Leukoc Biol ; 109(1): 257-273, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32991756

RESUMEN

Tuberculosis dates back to ancient times but it is not a problem of the past. Each year, millions of people die from tuberculosis. After inhalation of infectious droplet nuclei, Mycobacterium tuberculosis reaches the lungs where it can manipulate the immune system and survive within host macrophages, establishing a persistent infection. The signaling lymphocytic activation molecule family member 1 (SLAMF1) is a self-ligand receptor that can internalize gram-negative bacteria and regulate macrophages' phagosomal functions. In tuberculosis, SLAMF1 promotes Th1-protective responses. In this work, we studied the role of SLAMF1 on macrophages' functions during M. tuberculosis infection. Our results showed that both M. tuberculosis and IFN-γ stimulation induce SLAMF1 expression in macrophages from healthy donor and Tohoku Hospital Pediatrcs-1 cells. Costimulation through SLAMF1 with an agonistic antibody resulted in an enhanced internalization of M. tuberculosis by macrophages. Interestingly, we found that SLAMF1 interacts with M. tuberculosis and colocalizes with the bacteria and with early and late endosomes/lysosomes markers (EEA1 and LAMP2), suggesting that SLAMF1 recognize M. tuberculosis and participate in the endolysosomal maturation process. Notably, increased levels of SLAMF1 were detected in CD14 cells from pleural effusions of tuberculosis patients, indicating that SLAMF1 might have an active function at the site of infection. Taken together, our results provide evidence that SLAMF1 improves the uptake of M. tuberculosis by human monocyte-derived macrophages.


Asunto(s)
Macrófagos/inmunología , Fagocitosis/inmunología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Tuberculosis Pulmonar/inmunología , Adolescente , Adulto , Anciano , Endosomas/inmunología , Femenino , Humanos , Lisosomas/inmunología , Macrófagos/microbiología , Masculino , Persona de Mediana Edad , Mycobacterium tuberculosis/inmunología , Transducción de Señal/inmunología , Adulto Joven
17.
Eur J Immunol ; 39(9): 2450-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19714575

RESUMEN

Tuberculous pleurisy, one of the most common manifestations of extrapulmonary tuberculosis, is characterized by a T-cell-mediated hypersensitivity reaction along with a Th1 immune profile. In this study, we investigated functional cross-talk among T and NK cells in human tuberculous pleurisy. We found that endogenously activated pleural fluid-derived NK cells express high ICAM-1 levels and induce T-cell activation ex vivo through ICAM-1. Besides, upon in vitro stimulation with monokines and PAMP, resting peripheral blood NK cells increased ICAM-1 expression leading to cellular activation and Th1 polarization of autologous T cells. Furthermore, these effects were abolished by anti-ICAM-1 Ab. Hence, NK cells may contribute to the adaptive immune response by a direct cell-contact-dependent mechanism in the context of Mycobacterium tuberculosis infection.


Asunto(s)
Molécula 1 de Adhesión Intercelular/metabolismo , Células Asesinas Naturales/inmunología , Mycobacterium tuberculosis , Linfocitos T/inmunología , Tuberculosis Pleural/inmunología , Adulto , Antígeno CD11a/inmunología , Antígeno CD56/inmunología , Comunicación Celular/inmunología , Cisteína/análogos & derivados , Cisteína/farmacología , Humanos , Molécula 1 de Adhesión Intercelular/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/inmunología , Interleucina-12/farmacología , Interleucina-15/farmacología , Interleucina-18/farmacología , Lipopolisacáridos/farmacología , Lipoproteínas/farmacología , Persona de Mediana Edad , Fosfotransferasas (Aceptor del Grupo Fosfato)
18.
Immunol Cell Biol ; 88(7): 716-26, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20212510

RESUMEN

During a chronic infection such as tuberculosis, the pool of tissue dendritic cells (DC) must be renewed by recruitment of both circulating DC progenitors and monocytes (Mo). However, the microenvironment of the inflammatory site affects Mo differentiation. As DC are critical for initiating a Mycobacterium tuberculosis-specific T-cell response, we argue that interference of M. tuberculosis with a correct DC generation would signify a mechanism of immune evasion. In this study, we showed that early interaction of γ-irradiated M. tuberculosis with Mo subverts DC differentiation in vitro. We found that irradiated M. tuberculosis effect involves (1) the loss of a significant fraction of monocyte population and (2) an altered differentiation process of the surviving monocyte subpopulation. Moreover, in the absence of irradiated M. tuberculosis, DC consist in a major DC-specific intercellular adhesion molecule 3-grabbing non-integrin receptor (DC-SIGN(high))/CD86(low) and minor DC-SIGN(low)/CD86(high) subpopulations, whereas in the presence of bacteria, there is an enrichment of DC-SIGN(low)/CD86(high) population. Besides, this population enlarged by irradiated M. tuberculosis, which is characterized by a reduced CD1b expression, correlates with a reduced induction of specific T-lymphocyte proliferation. The loss of CD1molecules partially involves toll-like receptors (TLR-2)/p38 MAPK activation. Finally, several features of Mo, which have been differentiated into DC in the presence of irradiated M. tuberculosis, resemble the features of DC obtained from patients with active tuberculosis. In conclusion, we suggest that M. tuberculosis escapes from acquired immune response in tuberculosis may be caused by an altered differentiation into DC leading to a poor M. tuberculosis-specific T-cell response.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/microbiología , Adulto , Antígenos CD1/metabolismo , Antígeno B7-2/metabolismo , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Humanos , Interleucina-10/metabolismo , Lectinas Tipo C/metabolismo , Prueba de Cultivo Mixto de Linfocitos , Macrófagos/inmunología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Persona de Mediana Edad , Mycobacterium tuberculosis/patogenicidad , Receptores de Superficie Celular/metabolismo , Receptor Toll-Like 2/metabolismo , Tuberculosis/inmunología , Tuberculosis/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Front Immunol ; 11: 581911, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33679685

RESUMEN

The cholinergic system is present in both bacteria and mammals and regulates inflammation during bacterial respiratory infections through neuronal and non-neuronal production of acetylcholine (ACh) and its receptors. However, the presence of this system during the immunopathogenesis of pulmonary tuberculosis (TB) in vivo and in its causative agent Mycobacterium tuberculosis (Mtb) has not been studied. Therefore, we used an experimental model of progressive pulmonary TB in BALB/c mice to quantify pulmonary ACh using high-performance liquid chromatography during the course of the disease. In addition, we performed immunohistochemistry in lung tissue to determine the cellular expression of cholinergic system components, and then administered nicotinic receptor (nAChR) antagonists to validate their effect on lung bacterial burden, inflammation, and pro-inflammatory cytokines. Finally, we subjected Mtb cultures to colorimetric analysis to reveal the production of ACh and the effect of ACh and nAChR antagonists on Mtb growth. Our results show high concentrations of ACh and expression of its synthesizing enzyme choline acetyltransferase (ChAT) during early infection in lung epithelial cells and macrophages. During late progressive TB, lung ACh upregulation was even higher and coincided with ChAT and α7 nAChR subunit expression in immune cells. Moreover, the administration of nAChR antagonists increased pro-inflammatory cytokines, reduced bacillary loads and synergized with antibiotic therapy in multidrug resistant TB. Finally, in vitro studies revealed that the bacteria is capable of producing nanomolar concentrations of ACh in liquid culture. In addition, the administration of ACh and nicotinic antagonists to Mtb cultures induced or inhibited bacterial proliferation, respectively. These results suggest that Mtb possesses a cholinergic system and upregulates the lung non-neuronal cholinergic system, particularly during late progressive TB. The upregulation of the cholinergic system during infection could aid both bacterial growth and immunomodulation within the lung to favor disease progression. Furthermore, the therapeutic efficacy of modulating this system suggests that it could be a target for treating the disease.


Asunto(s)
Sistema Colinérgico no Neuronal/fisiología , Tuberculosis Pulmonar/metabolismo , Tuberculosis Pulmonar/patología , Acetilcolina/metabolismo , Animales , Colina O-Acetiltransferasa/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Inflamación/metabolismo , Inflamación/patología , Pulmón/metabolismo , Pulmón/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Antagonistas Nicotínicos/farmacología , Sistema Colinérgico no Neuronal/efectos de los fármacos , Receptores Nicotínicos/metabolismo , Regulación hacia Arriba/fisiología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
20.
Elife ; 92020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-32223897

RESUMEN

While tuberculosis (TB) is a risk factor in HIV-1-infected individuals, the mechanisms by which Mycobacterium tuberculosis (Mtb) worsens HIV-1 pathogenesis remain scarce. We showed that HIV-1 infection is exacerbated in macrophages exposed to TB-associated microenvironments due to tunneling nanotube (TNT) formation. To identify molecular factors associated with TNT function, we performed a transcriptomic analysis in these macrophages, and revealed the up-regulation of Siglec-1 receptor. Siglec-1 expression depends on Mtb-induced production of type I interferon (IFN-I). In co-infected non-human primates, Siglec-1 is highly expressed by alveolar macrophages, whose abundance correlates with pathology and activation of IFN-I/STAT1 pathway. Siglec-1 localizes mainly on microtubule-containing TNT that are long and carry HIV-1 cargo. Siglec-1 depletion decreases TNT length, diminishes HIV-1 capture and cell-to-cell transfer, and abrogates the exacerbation of HIV-1 infection induced by Mtb. Altogether, we uncover a deleterious role for Siglec-1 in TB-HIV-1 co-infection and open new avenues to understand TNT biology.


Asunto(s)
VIH-1/patogenicidad , Interferón Tipo I/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/virología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Tuberculosis Pulmonar/inmunología , Animales , Células Cultivadas , Coinfección/inmunología , Femenino , Perfilación de la Expresión Génica , Infecciones por VIH , Humanos , Macaca mulatta , Masculino , Nanotubos , Lectina 1 Similar a Ig de Unión al Ácido Siálico/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA