Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Elife ; 122024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922679

ABSTRACT

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.


Subject(s)
Cell Movement , Dendritic Cells , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Monocytes , Mycobacterium tuberculosis , Tuberculosis , Dendritic Cells/metabolism , Dendritic Cells/immunology , Monocytes/metabolism , Monocytes/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mycobacterium tuberculosis/immunology , Animals , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Mice , Toll-Like Receptor 2/metabolism , Mice, Inbred C57BL , Female
2.
PLoS Negl Trop Dis ; 16(11): e0010950, 2022 11.
Article in English | MEDLINE | ID: mdl-36441810

ABSTRACT

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.


Subject(s)
Brucella abortus , RNA , Humans , Brucella abortus/genetics
4.
J Leukoc Biol ; 112(1): 173-184, 2022 07.
Article in English | MEDLINE | ID: mdl-34811790

ABSTRACT

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.


Subject(s)
Macrophage Activation , Whooping Cough , Bordetella pertussis , Humans , Interleukin-6/metabolism , Macrophages/metabolism , Tumor Necrosis Factor-alpha/metabolism , Whooping Cough/metabolism
5.
J Leukoc Biol ; 109(1): 257-273, 2021 01.
Article in English | MEDLINE | ID: mdl-32991756

ABSTRACT

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.


Subject(s)
Macrophages/immunology , Phagocytosis/immunology , Signaling Lymphocytic Activation Molecule Family Member 1/immunology , Tuberculosis, Pulmonary/immunology , Adolescent , Adult , Aged , Endosomes/immunology , Female , Humans , Lysosomes/immunology , Macrophages/microbiology , Male , Middle Aged , Mycobacterium tuberculosis/immunology , Signal Transduction/immunology , Young Adult
6.
Cell Rep ; 33(13): 108547, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378679

ABSTRACT

Mycobacterium tuberculosis (Mtb) regulates the macrophage metabolic state to thrive in the host, yet the responsible mechanisms remain elusive. Macrophage activation toward the microbicidal (M1) program depends on the HIF-1α-mediated metabolic shift from oxidative phosphorylation (OXPHOS) toward glycolysis. Here, we ask whether a tuberculosis (TB) microenvironment changes the M1 macrophage metabolic state. We expose M1 macrophages to the acellular fraction of tuberculous pleural effusions (TB-PEs) and find lower glycolytic activity, accompanied by elevated levels of OXPHOS and bacillary load, compared to controls. The eicosanoid fraction of TB-PE drives these metabolic alterations. HIF-1α stabilization reverts the effect of TB-PE by restoring M1 metabolism. Furthermore, Mtb-infected mice with stabilized HIF-1α display lower bacillary loads and a pronounced M1-like metabolic profile in alveolar macrophages (AMs). Collectively, we demonstrate that lipids from a TB-associated microenvironment alter the M1 macrophage metabolic reprogramming by hampering HIF-1α functions, thereby impairing control of Mtb infection.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipids/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mycobacterium tuberculosis/metabolism , Tuberculosis, Pleural/metabolism , Animals , Bacterial Load , Eicosanoids/pharmacology , Female , Glycolysis/drug effects , Host-Pathogen Interactions , Humans , Macrophage Activation , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Pleural Effusion , Tuberculosis, Pleural/microbiology
7.
PLoS Pathog ; 16(10): e1008929, 2020 10.
Article in English | MEDLINE | ID: mdl-33002063

ABSTRACT

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.


Subject(s)
Foam Cells/microbiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipid Metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/pathogenicity , Animals , Lipid Droplets/metabolism , Macrophage Activation/physiology , Macrophages/metabolism , Male , Mice, Inbred BALB C , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology
9.
Elife ; 92020 03 30.
Article in English | MEDLINE | ID: mdl-32223897

ABSTRACT

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.


Subject(s)
HIV-1/pathogenicity , Interferon Type I/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Sialic Acid Binding Ig-like Lectin 1/genetics , Tuberculosis, Pulmonary/immunology , Animals , Cells, Cultured , Coinfection/immunology , Female , Gene Expression Profiling , HIV Infections , Humans , Macaca mulatta , Male , Nanotubes , Sialic Acid Binding Ig-like Lectin 1/immunology
10.
Front Immunol ; 11: 581911, 2020.
Article in English | MEDLINE | ID: mdl-33679685

ABSTRACT

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.


Subject(s)
Non-Neuronal Cholinergic System/physiology , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/pathology , Acetylcholine/metabolism , Animals , Choline O-Acetyltransferase/metabolism , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammation/metabolism , Inflammation/pathology , Lung/metabolism , Lung/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/drug effects , Neurons/metabolism , Neurons/pathology , Nicotinic Antagonists/pharmacology , Non-Neuronal Cholinergic System/drug effects , Receptors, Nicotinic/metabolism , Up-Regulation/physiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
11.
Front Immunol ; 10: 2181, 2019.
Article in English | MEDLINE | ID: mdl-31572389

ABSTRACT

Brucella abortus, the causative agent of brucellosis, displays many resources to evade T cell responses conducive to persist inside the host. Our laboratory has previously showed that infection of human monocytes with B. abortus down-modulates the IFN-γ-induced MHC-II expression. Brucella outer membrane lipoproteins are structural components involved in this phenomenon. Moreover, IL-6 is the soluble factor that mediated MHC-II down-regulation. Yet, the MHC-II down-regulation exerted by lipoproteins was less marked than the one observed as consequence of infection. This led us to postulate that there should be other components associated with viable bacteria that may act together with lipoproteins in order to diminish MHC-II. Our group has recently demonstrated that B. abortus RNA (PAMP related to pathogens' viability or vita-PAMP) is involved in MHC-I down-regulation. Therefore, in this study we investigated if B. abortus RNA could be contributing to the down-regulation of MHC-II. This PAMP significantly down-modulated the IFN-γ-induced MHC-II surface expression on THP-1 cells as well as in primary human monocytes and murine bone marrow macrophages. The expression of other molecules up-regulated by IFN-γ (such as co-stimulatory molecules) was stimulated on monocytes treated with B. abortus RNA. This result shows that this PAMP does not alter all IFN-γ-induced molecules globally. We also showed that other bacterial and parasitic RNAs caused MHC-II surface expression down-modulation indicating that this phenomenon is not restricted to B. abortus. Moreover, completely degraded RNA was also able to reproduce the phenomenon. MHC-II down-regulation on monocytes treated with RNA and L-Omp19 (a prototypical lipoprotein of B. abortus) was more pronounced than in monocytes stimulated with both components separately. We also demonstrated that B. abortus RNA along with its lipoproteins decrease MHC-II surface expression predominantly by a mechanism of inhibition of MHC-II expression. Regarding the signaling pathway, we demonstrated that IL-6 is a soluble factor implicated in B. abortus RNA and lipoproteins-triggered MHC-II surface down-regulation. Finally, CD4+ T cells functionality was affected as macrophages treated with these components showed lower antigen presentation capacity. Therefore, B. abortus RNA and lipoproteins are two PAMPs that contribute to MHC-II down-regulation on monocytes/macrophages diminishing CD4+ T cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Macrophages/immunology , Monocytes/immunology , RNA, Bacterial/immunology , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Brucella abortus/genetics , Brucella abortus/immunology , Brucella abortus/physiology , Brucellosis/immunology , Brucellosis/microbiology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Down-Regulation/immunology , Female , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lipoproteins/immunology , Lipoproteins/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , RNA, Bacterial/genetics , THP-1 Cells
13.
Cell Rep ; 26(13): 3586-3599.e7, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30917314

ABSTRACT

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.


Subject(s)
HIV Infections/complications , Interleukin-10/metabolism , Macrophages/pathology , Nanotubes , STAT3 Transcription Factor/metabolism , Tuberculosis, Pulmonary/complications , Adult , Aged , Animals , Cells, Cultured , Coinfection/pathology , Coinfection/virology , Female , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , Humans , Macaca mulatta , Macrophage Activation , Macrophages/virology , Male , Middle Aged , Mycobacterium tuberculosis , Signal Transduction , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Virus Replication , Young Adult
14.
Sci Rep ; 9(1): 187, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30655556

ABSTRACT

CD8+T cells contribute to tuberculosis (TB) infection control by inducing death of infected macrophages. Mycobacterium tuberculosis (Mtb) infection is associated with increased PD-1/PD-L1 expression and alternative activation of macrophages. We aimed to study the role of PD-1 pathway and macrophage polarization on Mtb-specific CD8+T cell-induced macrophage death. We observed that both PD-L1 on CD14+ cells and PD-1 on CD8+T cells were highly expressed at the site of infection in pleurisy TB patients' effusion samples (PEMC). Moreover, a significant increase in CD8+T cells' Mtb-specific degranulation from TB-PEMC vs. TB-PBMC was observed, which correlated with PD-1 and PDL-1 expression. In an in vitro model, M1 macrophages were more susceptible to Mtb-specific CD8+T cells' cytotoxicity compared to M2a macrophages and involved the transfer of cytolytic effector molecules from CD8+T lymphocytes to target cells. Additionally, PD-L1 blocking significantly increased the in vitro Ag-specific CD8+T cell cytotoxicity against IFN-γ-activated macrophages but had no effect over cytotoxicity on IL-4 or IL-10-activated macrophages. Interestingly, PD-L1 blocking enhanced Mtb-specific CD8+ T cell killing of CD14+ cells from human tuberculous pleural effusion samples. Our data indicate that PD-1/PD-L1 pathway modulates antigen-specific cytotoxicity against M1 targets in-vitro and encourage the exploration of checkpoint blockade as new adjuvant for TB therapies.


Subject(s)
B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Death , Macrophages/microbiology , Mycobacterium tuberculosis/immunology , Programmed Cell Death 1 Receptor/metabolism , Blood Specimen Collection , CD8-Positive T-Lymphocytes/microbiology , Humans , Macrophages/pathology , Pleural Effusion/microbiology , T-Lymphocytes, Cytotoxic/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control
16.
Front Immunol ; 9: 1123, 2018.
Article in English | MEDLINE | ID: mdl-29946317

ABSTRACT

DC-SIGN (CD209/CLEC4L) is a C-type lectin receptor (CLR) that serves as a reliable cell-surface marker of interleukin 4 (IL-4)-activated human macrophages [M(IL-4)], which historically represent the most studied subset within the M2 spectrum of macrophage activation. Although DC-SIGN plays important roles in Mycobacterium tuberculosis (Mtb) interactions with dendritic cells, its contribution to the Mtb-macrophage interaction remains poorly understood. Since high levels of IL-4 are correlated with tuberculosis (TB) susceptibility and progression, we investigated the role of DC-SIGN in M(IL-4) macrophages in the TB context. First, we demonstrate that DC-SIGN expression is present both in CD68+ macrophages found in tuberculous pulmonary lesions of non-human primates, and in the CD14+ cell population isolated from pleural effusions obtained from TB patients (TB-PE). Likewise, we show that DC-SIGN expression is accentuated in M(IL-4) macrophages derived from peripheral blood CD14+ monocytes isolated from TB patients, or in macrophages stimulated with acellular TB-PE, arguing for the pertinence of DC-SIGN-expressing macrophages in TB. Second, using a siRNA-mediated gene silencing approach, we performed a transcriptomic analysis of DC-SIGN-depleted M(IL-4) macrophages and revealed the upregulation of pro-inflammatory signals in response to challenge with Mtb, as compared to control cells. This pro-inflammatory gene signature was confirmed by RT-qPCR, cytokine/chemokine-based protein array, and ELISA analyses. We also found that inactivation of DC-SIGN renders M(IL-4) macrophages less permissive to Mtb intracellular growth compared to control cells, despite the equal level of bacteria uptake. Last, at the molecular level, we show that DC-SIGN interferes negatively with the pro-inflammatory response and control of Mtb intracellular growth mediated by another CLR, Dectin-1 (CLEC7A). Collectively, this study highlights a dual role for DC-SIGN as, on the one hand, being a host factor granting advantage for Mtb to parasitize macrophages and, on the other hand, representing a molecular switch to turn off the pro-inflammatory response in these cells to prevent potential immunopathology associated to TB.


Subject(s)
Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Macrophages/immunology , Macrophages/metabolism , Mycobacterium tuberculosis/immunology , Receptors, Cell Surface/metabolism , Tuberculosis/immunology , Tuberculosis/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Survival/genetics , Cell Survival/immunology , Cytokines/metabolism , Female , Gene Expression , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators/metabolism , Lectins, C-Type/genetics , Macaca mulatta , Macrophages/microbiology , Monocytes/immunology , Monocytes/metabolism , Phagocytosis/immunology , Receptors, Cell Surface/genetics , Tuberculosis/genetics , Tuberculosis/microbiology
17.
Front Immunol ; 9: 459, 2018.
Article in English | MEDLINE | ID: mdl-29593722

ABSTRACT

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.


Subject(s)
Acetyl-CoA C-Acetyltransferase/immunology , Gene Expression Regulation, Enzymologic/immunology , Interleukin-10/immunology , Mycobacterium tuberculosis/immunology , Pleural Effusion/immunology , STAT3 Transcription Factor/immunology , Sterol O-Acyltransferase , Tuberculosis, Pleural/immunology , Up-Regulation/immunology , Acetyl-CoA C-Acetyltransferase/genetics , Animals , Female , Foam Cells , Humans , Interleukin-10/genetics , Male , Mice , Mice, Knockout , Mycobacterium tuberculosis/genetics , Pleural Effusion/genetics , Pleural Effusion/pathology , STAT3 Transcription Factor/genetics , Tuberculosis, Pleural/genetics , Tuberculosis, Pleural/pathology
18.
PLoS Pathog ; 13(8): e1006527, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28767704

ABSTRACT

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.


Subject(s)
Brucellosis/immunology , ErbB Receptors/immunology , Immune Evasion/immunology , Monocytes/immunology , RNA, Bacterial/immunology , Toll-Like Receptor 8/immunology , Animals , Brucella abortus/immunology , Cross-Priming/immunology , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Histocompatibility Antigens Class I/biosynthesis , Humans , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Monocytes/microbiology , Signal Transduction/immunology
19.
Mediators Inflamm ; 2017: 2810606, 2017.
Article in English | MEDLINE | ID: mdl-28852268

ABSTRACT

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.


Subject(s)
Interleukin-8/metabolism , Mycobacterium tuberculosis/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Chemokines/metabolism , Chemotaxis/drug effects , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Flow Cytometry , Humans , Immunity, Innate/drug effects , Phagocytosis/drug effects
20.
Tuberculosis (Edinb) ; 103: 16-23, 2017 03.
Article in English | MEDLINE | ID: mdl-28237029

ABSTRACT

C5a anaphylatoxin is a component of the complement system involved in the modulation of T-cell polarization. Herein we investigated whether C5a receptors, C5aR and C5L2, modulate the cytokine profiles induced by Mycobacterium tuberculosis (Mtb). We analyzed the impact of both receptors on T helper cell polarization induced by the multidrug resistant outbreak strain named M, which is a poor IFN-γ inducer compared with the laboratory strain H37Rv. To this aim, we first blocked C5aR or C5L2 of peripheral blood monocytes (Mo) from patients with tuberculosis and healthy donors, then we stimulated the Mo either with H37Rv or the M strain, and finally we analyzed cytokine profiles of Mo/macrophages (MΦ) and CD4+ T-cells. We found that: (i) Mtb modulated the expression of both C5a receptors, (ii) C5aR inhibited the expansion of CD4+IFN-γ+ lymphocytes stimulated by the M strain but not by H37Rv, (iii) both receptors modulated the Mo/MΦ cytokine expression induced by Mtb. We conclude that C5aR, but not C5L2, plays a role in T helper cell polarization induced by Mtb and that this effect is strain- and donor-dependent. We speculate that the epidemiologically successful M strain takes advantage of this C5aR-mediated inhibition of Th1 polarization to survive within the host.


Subject(s)
Cytokines/immunology , Disease Outbreaks , Mycobacterium tuberculosis/immunology , Receptor, Anaphylatoxin C5a/immunology , Th1 Cells/immunology , Tuberculosis, Multidrug-Resistant/immunology , Adolescent , Adult , Aged , Case-Control Studies , Cells, Cultured , Cytokines/metabolism , Female , Genotype , Host-Pathogen Interactions , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Phenotype , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Th1 Cells/metabolism , Th1 Cells/microbiology , Time Factors , Tuberculosis, Multidrug-Resistant/metabolism , Tuberculosis, Multidrug-Resistant/microbiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...