Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 617(7960): 386-394, 2023 May.
Article in English | MEDLINE | ID: mdl-37100912

ABSTRACT

Inflammation is a complex physiological process triggered in response to harmful stimuli1. It involves cells of the immune system capable of clearing sources of injury and damaged tissues. Excessive inflammation can occur as a result of infection and is a hallmark of several diseases2-4. The molecular bases underlying inflammatory responses are not fully understood. Here we show that the cell surface glycoprotein CD44, which marks the acquisition of distinct cell phenotypes in the context of development, immunity and cancer progression, mediates the uptake of metals including copper. We identify a pool of chemically reactive copper(II) in mitochondria of inflammatory macrophages that catalyses NAD(H) redox cycling by activating hydrogen peroxide. Maintenance of NAD+ enables metabolic and epigenetic programming towards the inflammatory state. Targeting mitochondrial copper(II) with supformin (LCC-12), a rationally designed dimer of metformin, induces a reduction of the NAD(H) pool, leading to metabolic and epigenetic states that oppose macrophage activation. LCC-12 interferes with cell plasticity in other settings and reduces inflammation in mouse models of bacterial and viral infections. Our work highlights the central role of copper as a regulator of cell plasticity and unveils a therapeutic strategy based on metabolic reprogramming and the control of epigenetic cell states.


Subject(s)
Cell Plasticity , Copper , Inflammation , Signal Transduction , Animals , Mice , Copper/metabolism , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , NAD/metabolism , Signal Transduction/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Hydrogen Peroxide/metabolism , Epigenesis, Genetic/drug effects , Metformin/analogs & derivatives , Oxidation-Reduction , Cell Plasticity/drug effects , Cell Plasticity/genetics , Macrophage Activation/drug effects , Macrophage Activation/genetics
2.
PLoS Pathog ; 19(3): e1011192, 2023 03.
Article in English | MEDLINE | ID: mdl-36888688

ABSTRACT

Progression of tuberculosis is tightly linked to a disordered immune balance, resulting in inability of the host to restrict intracellular bacterial replication and its subsequent dissemination. The immune response is mainly characterized by an orchestrated recruitment of inflammatory cells secreting cytokines. This response results from the activation of innate immunity receptors that trigger downstream intracellular signaling pathways involving adaptor proteins such as the TIR-containing adaptor protein (Tirap). In humans, resistance to tuberculosis is associated with a loss-of-function in Tirap. Here, we explore how genetic deficiency in Tirap impacts resistance to Mycobacterium tuberculosis (Mtb) infection in a mouse model and ex vivo. Interestingly, compared to wild type littermates, Tirap heterozygous mice were more resistant to Mtb infection. Upon investigation at the cellular level, we observed that mycobacteria were not able to replicate in Tirap-deficient macrophages compared to wild type counterparts. We next showed that Mtb infection induced Tirap expression which prevented phagosomal acidification and rupture. We further demonstrate that the Tirap-mediated anti-tuberculosis effect occurs through a Cish-dependent signaling pathway. Our findings provide new molecular evidence about how Mtb manipulates innate immune signaling to enable intracellular replication and survival of the pathogen, thus paving the way for host-directed approaches to treat tuberculosis.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Mice , Animals , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Hydrogen-Ion Concentration , Membrane Glycoproteins/metabolism
3.
PLoS Pathog ; 18(5): e1010498, 2022 05.
Article in English | MEDLINE | ID: mdl-35587469

ABSTRACT

Drug repurposing has the advantage of shortening regulatory preclinical development steps. Here, we screened a library of drug compounds, already registered in one or several geographical areas, to identify those exhibiting antiviral activity against SARS-CoV-2 with relevant potency. Of the 1,942 compounds tested, 21 exhibited a substantial antiviral activity in Vero-81 cells. Among them, clofoctol, an antibacterial drug used for the treatment of bacterial respiratory tract infections, was further investigated due to its favorable safety profile and pharmacokinetic properties. Notably, the peak concentration of clofoctol that can be achieved in human lungs is more than 20 times higher than its IC50 measured against SARS-CoV-2 in human pulmonary cells. This compound inhibits SARS-CoV-2 at a post-entry step. Lastly, therapeutic treatment of human ACE2 receptor transgenic mice decreased viral load, reduced inflammatory gene expression and lowered pulmonary pathology. Altogether, these data strongly support clofoctol as a therapeutic candidate for the treatment of COVID-19 patients.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Animals , Antiviral Agents/pharmacology , Chlorobenzenes , Chlorocebus aethiops , Cresols , Humans , Lung , Mice , Vero Cells
4.
Brain Behav Immun ; 117: 20-35, 2024 03.
Article in English | MEDLINE | ID: mdl-38157948

ABSTRACT

BACKGROUND: Cerebral malaria (CM) is a fatal neuroinflammatory syndrome caused (in humans) by the protozoa Plasmodium (P.) falciparum. Glial cell activation is one of the mechanisms that contributes to neuroinflammation in CM. RESULT: By studying a mouse model of CM (caused by P. berghei ANKA), we describe that the induction of autophagy promoted p21-dependent senescence in astrocytes and that CXCL-10 was part of the senescence-associated secretory phenotype. Furthermore, p21 expression was observed in post-mortem brain and peripheral blood samples from patients with CM. Lastly, we found that the depletion of senescent astrocytes with senolytic drugs abrogated inflammation and protected mice from CM. CONCLUSION: Our data provide evidence for a novel mechanism through which astrocytes could be involved in the neuropathophysiology of CM. p21 gene expression in blood cell and an elevated plasma CXCL-10 concentration could be valuable biomarkers of CM in humans. In the end, we believe senolytic drugs shall open up new avenues to develop newer treatment options.


Subject(s)
Malaria, Cerebral , Humans , Animals , Mice , Neuroinflammatory Diseases , Astrocytes , Senotherapeutics , Autophagy
5.
Bioconjug Chem ; 34(9): 1563-1575, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37694903

ABSTRACT

Pneumococcal conjugate vaccines offer an excellent safety profile and high protection against the serotypes comprised in the vaccine. However, inclusion of protein antigens fromStreptococcus pneumoniaecombined with potent adjuvants and a suitable delivery system are expected to both extend protection to serotype strains not represented in the formulation and stimulate a broader immune response, thus more effective in young children, elderly, and immunocompromised populations. Along this line, nanoparticle (NP) delivery systems can enhance the immunogenicity of antigens by protecting them from degradation and increasing their uptake by antigen-presenting cells, as well as offering co-delivery with adjuvants. We report herein the encapsulation of a semisynthetic glycoconjugate (GC) composed of a synthetic tetrasaccharide mimicking theS. pneumoniae serotype 14 capsular polysaccharide (CP14) linked to the Pneumococcal surface protein A (PsaA) using chitosan NPs (CNPs). These GC-loaded chitosan nanoparticles (GC-CNPs) were not toxic to human monocyte-derived dendritic cells (MoDCs), showed enhanced uptake, and displayed better immunostimulatory properties in comparison to the naked GC. A comparative study was carried out in mice to evaluate the immune response elicited by the glycoconjugate-administered subcutaneously (SC), where the GC-CNPs displayed 100-fold higher IgG response as compared with the group treated with nonencapsulated GC. Overall, the study demonstrates the potential of this chitosan-based nanovaccine for efficient delivery of glycoconjugate antigens.


Subject(s)
Chitosan , Child , Aged , Humans , Animals , Mice , Child, Preschool , Pneumococcal Vaccines , Streptococcus pneumoniae , Adjuvants, Immunologic , Glycoconjugates/therapeutic use
6.
Antimicrob Agents Chemother ; 66(2): e0158121, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34807755

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak has highlighted the need for broad-spectrum antivirals against coronaviruses (CoVs). Here, pheophorbide a (Pba) was identified as a highly active antiviral molecule against human CoV-229E after bioguided fractionation of plant extracts. The antiviral activity of Pba was subsequently shown for SARS-CoV-2 and Middle East respiratory syndrome coronavirus (MERS-CoV), and its mechanism of action was further assessed, showing that Pba is an inhibitor of coronavirus entry by directly targeting the viral particle. Interestingly, the antiviral activity of Pba depends on light exposure, and Pba was shown to inhibit virus-cell fusion by stiffening the viral membrane, as demonstrated by cryoelectron microscopy. Moreover, Pba was shown to be broadly active against several other enveloped viruses and reduced SARS-CoV-2 and MERS-CoV replication in primary human bronchial epithelial cells. Pba is the first described natural antiviral against SARS-CoV-2 with direct photosensitive virucidal activity that holds potential for COVID-19 therapy or disinfection of SARS-CoV-2-contaminated surfaces.


Subject(s)
Biological Products , COVID-19 , Middle East Respiratory Syndrome Coronavirus , Antiviral Agents/pharmacology , Biological Products/pharmacology , Cryoelectron Microscopy , Humans , SARS-CoV-2
7.
PLoS Biol ; 17(3): e3000169, 2019 03.
Article in English | MEDLINE | ID: mdl-30822302

ABSTRACT

CD1d-restricted invariant natural killer T (iNKT) cells represent a heterogeneous population of lipid-reactive T cells that are involved in many immune responses, mediated through T-cell receptor (TCR)-dependent and/or independent activation. Although numerous microbial lipid antigens (Ags) have been identified, several lines of evidence have suggested the existence of relevant Ags of endogenous origin. However, the identification of their precise nature as well as the molecular mechanisms involved in their generation are still highly controversial and ill defined. Here, we identified two mammalian gangliosides-namely monosialoganglioside GM3 and disialoganglioside GD3-as endogenous activators for mouse iNKT cells. These glycosphingolipids are found in Toll-like receptor-stimulated dendritic cells (DC) as several species varying in their N-acyl fatty chain composition. Interestingly, their ability to activate iNKT cells is highly dependent on the ceramide backbone structure. Thus, both synthetic GM3 and GD3 comprising a d18:1-C24:1 ceramide backbone were able to activate iNKT cells in a CD1d-dependent manner. GM3 and GD3 are not directly recognized by the iNKT TCR and required the Ag presenting cell intracellular machinery to reveal their antigenicity. We propose a new concept in which iNKT cells can rapidly respond to pre-existing self-molecules after stress-induced structural changes in CD1d-expressing cells. Moreover, these gangliosides conferred partial protection in the context of bacterial infection. Thus, this report identified new biologically relevant lipid self-Ags for iNKT cells.


Subject(s)
Ceramides/metabolism , Gangliosides/metabolism , Natural Killer T-Cells/metabolism , Toll-Like Receptor 9/metabolism , Animals , Antigens, CD1d/metabolism , Bone Marrow Cells/metabolism , Dendritic Cells/metabolism , G(M3) Ganglioside/metabolism , Glycosphingolipids/metabolism , Male , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
8.
Int J Mol Sci ; 23(11)2022 May 24.
Article in English | MEDLINE | ID: mdl-35682578

ABSTRACT

Non-conventional T cells, such as γδ T and invariant natural killer T (iNKT) cells, are emerging players in fighting cancer. Alpha-galactosylceramide (α-GalCer) is used as an exogenous ligand to activate iNKT cells. Human cells don't have a direct pathway producing α-GalCer, which, however, can be produced by bacteria. We searched the literature for bacteria strains that are able to produce α-GalCer and used available sequencing data to analyze their presence in human tumor tissues and their association with survival. The modulatory effect of antibiotics on the concentration of α-GalCer was analyzed in mice. The human gut bacteria Bacteroides fragilis, Bacteroides vulgatus, and Prevotella copri produce α-GalCer structures that are able to activate iNKT cells. In mice, α-GalCer was depleted upon treatment with vancomycin. The three species were detected in colon adenocarcinoma (COAD) and rectum adenocarcinoma tissues, and Prevotella copri was also detected in bone tumors and glioblastoma tissues. Bacteroides vulgatus in COAD tissues correlated with better survival. In conclusion, α-GalCer-producing bacteria are part of the human gut microbiome and can infiltrate tumor tissues. These results suggest a new mechanism of interaction between bacteria and immune cells: α-GalCer produced by bacteria may activate non-conventional T cells in tumor tissues, where they can exert a direct or indirect anti-tumor activity.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Natural Killer T-Cells , Adenocarcinoma/metabolism , Animals , Bacteroides , Colonic Neoplasms/metabolism , Galactosylceramides , Immunotherapy , Mice , Mice, Inbred C57BL , Prevotella
9.
Infect Immun ; 89(9): e0018821, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34097474

ABSTRACT

Short-chain fatty acids (SCFAs) are the main metabolites produced by the gut microbiota via the fermentation of complex carbohydrates and fibers. Evidence suggests that SCFAs play a role in the control of infections through direct action both on microorganisms and on host signaling. This review summarizes the main microbicidal effects of SCFAs and discusses studies highlighting the effect of SCFAs in the virulence and viability of microorganisms. We also describe the diverse and complex modes of action of the SCFAs on the immune system in the face of infections with a specific focus on bacterial and viral respiratory infections. A growing body of evidence suggests that SCFAs protect against lung infections. Finally, we present potential strategies that may be leveraged to exploit the biological properties of SCFAs for increasing effectiveness and optimizing patient benefits.


Subject(s)
Anti-Infective Agents/therapeutic use , Fatty Acids, Volatile/therapeutic use , Infections/drug therapy , Lung/drug effects , Animals , Anti-Infective Agents/immunology , Anti-Infective Agents/metabolism , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Humans , Infections/immunology , Infections/microbiology , Lung/immunology , Lung/microbiology , Lung/virology , Microbial Viability , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Signal Transduction/immunology , Virulence
10.
Infect Immun ; 89(9): e0073420, 2021 08 16.
Article in English | MEDLINE | ID: mdl-33820816

ABSTRACT

Along with respiratory tract disease per se, viral respiratory infections can also cause extrapulmonary complications with a potentially critical impact on health. In the present study, we used an experimental model of influenza A virus (IAV) infection to investigate the nature and outcome of the associated gut disorders. In IAV-infected mice, the signs of intestinal injury and inflammation, altered gene expression, and compromised intestinal barrier functions peaked on day 7 postinfection. As a likely result of bacterial component translocation, gene expression of inflammatory markers was upregulated in the liver. These changes occurred concomitantly with an alteration of the composition of the gut microbiota and with a decreased production of the fermentative, gut microbiota-derived products short-chain fatty acids (SCFAs). Gut inflammation and barrier dysfunction during influenza were not attributed to reduced food consumption, which caused in part gut dysbiosis. Treatment of IAV-infected mice with SCFAs was associated with an enhancement of intestinal barrier properties, as assessed by a reduction in the translocation of dextran and a decrease in inflammatory gene expression in the liver. Lastly, SCFA supplementation during influenza tended to reduce the translocation of the enteric pathogen Salmonella enterica serovar Typhimurium and to enhance the survival of doubly infected animals. Collectively, influenza virus infection can remotely impair the gut's barrier properties and trigger secondary enteric infections. The latter phenomenon can be partially countered by SCFA supplementation.


Subject(s)
Enterobacteriaceae Infections/etiology , Fatty Acids, Volatile/biosynthesis , Host-Pathogen Interactions , Influenza A virus/physiology , Influenza, Human/complications , Influenza, Human/virology , Intestinal Mucosa/metabolism , Microbial Interactions , Disease Susceptibility , Dysbiosis , Enterobacteriaceae Infections/metabolism , Host-Pathogen Interactions/immunology , Humans , Influenza, Human/metabolism , Intestinal Mucosa/immunology
11.
PLoS Pathog ; 14(10): e1007360, 2018 10.
Article in English | MEDLINE | ID: mdl-30372491

ABSTRACT

Secondary bacterial infections contribute to the excess morbidity and mortality of influenza A virus (IAV) infection. Disruption of lung integrity and impaired antibacterial immunity during IAV infection participate in colonization and dissemination of the bacteria out of the lungs. One key feature of IAV infection is the profound alteration of lung myeloid cells, characterized by the recruitment of deleterious inflammatory monocytes. We herein report that IAV infection causes a transient decrease of lung conventional dendritic cells (cDCs) (both cDC1 and cDC2) peaking at day 7 post-infection. While triggering emergency monopoiesis, IAV transiently altered the differentiation of cDCs in the bone marrow, the cDC1-biaised pre-DCs being particularly affected. The impaired cDC differentiation during IAV infection was independent of type I interferons (IFNs), IFN-γ, TNFα and IL-6 and was not due to an intrinsic dysfunction of cDC precursors. The alteration of cDC differentiation was associated with a drop of local and systemic production of Fms-like tyrosine kinase 3 ligand (Flt3-L), a critical cDC differentiation factor. Overexpression of Flt3-L during IAV infection boosted the cDC progenitors' production in the BM, replenished cDCs in the lungs, decreased inflammatory monocytes' infiltration and lowered lung damages. This was associated with partial protection against secondary pneumococcal infection, as reflected by reduced bacterial dissemination and prolonged survival. These findings highlight the impact of distal viral infection on cDC genesis in the BM and suggest that Flt3-L may have potential applications in the control of secondary infections.


Subject(s)
Dendritic Cells/immunology , Influenza A virus/immunology , Lung/immunology , Membrane Proteins/immunology , Orthomyxoviridae Infections/virology , Pneumococcal Infections/immunology , Superinfection/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/microbiology , Dendritic Cells/virology , Lung/microbiology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/complications , Pneumococcal Infections/microbiology , Pneumococcal Infections/virology , Receptor, Interferon alpha-beta/physiology , Streptococcus pneumoniae/immunology
12.
J Lipid Res ; 60(11): 1892-1904, 2019 11.
Article in English | MEDLINE | ID: mdl-31484693

ABSTRACT

The glycosphingolipid, α-galactosylceramide (αGalCer), when presented by CD1d on antigen-presenting cells, efficiently activates invariant natural killer T (iNKT) cells. Thereby, it modulates immune responses against tumors, microbial and viral infections, and autoimmune diseases. Recently, the production of αGalCer by Bacteroidetes from the human gut microbiome was elucidated. Using hydrophilic interaction chromatography coupled to MS2, we screened murine intestinal tracts to identify and quantify αGalCers, and we investigated the αGalCer response to different dietary and physiologic conditions. In both the cecum and the colon of mice, we found 1-15 pmol of αGalCer per milligram of protein; in contrast, mice lacking microbiota (germ-free mice) and fed identical diet did not harbor αGalCer. The identified αGalCer contained a ß(R)-hydroxylated hexadecanoyl chain N-linked to C18-sphinganine, which differed from what has been reported with Bacteroides fragilis Unlike ß-anomeric structures, but similar to αGalCers from B. fragilis, the synthetic form of the murine αGalCer induced iNKT cell activation in vitro. Last, we observed a decrease in αGalCer production in mice exposed to conditions that alter the composition of the gut microbiota, including Western type diet, colitis, and influenza A virus infection. Collectively, this study suggests that αGalCer is produced by commensals in the mouse intestine and reveals that stressful conditions causing dysbiosis alter its synthesis. The consequences of this altered production on iNKT cell-mediated local and systemic immune responses are worthy of future studies.


Subject(s)
Bacteroides fragilis/chemistry , Bacteroides fragilis/immunology , Diet , Galactosylceramides/immunology , Inflammation/immunology , Intestine, Large/immunology , Intestine, Large/metabolism , Animals , Galactosylceramides/genetics , Inflammation/microbiology , Intestine, Large/microbiology , Mice , Mice, Inbred Strains
13.
Infect Immun ; 86(7)2018 07.
Article in English | MEDLINE | ID: mdl-29661933

ABSTRACT

Severe bacterial (pneumococcal) infections are commonly associated with influenza and are significant contributors to the excess morbidity and mortality of influenza. Disruption of lung tissue integrity during influenza participates in bacterial pulmonary colonization and dissemination out of the lungs. Interleukin-22 (IL-22) has gained considerable interest in anti-inflammatory and anti-infection immunotherapy over the last decade. In the current study, we investigated the effect of exogenous IL-22 delivery on the outcome of pneumococcal superinfection postinfluenza. Our data show that exogenous treatment of influenza virus-infected mice with recombinant IL-22 reduces bacterial dissemination out of the lungs but is without effect on pulmonary bacterial burden. Reduced systemic bacterial dissemination was linked to reinforced pulmonary barrier functions, as revealed by total protein measurement in the bronchoalveolar fluids, intratracheal fluorescein isothiocyanate-dextran tracking, and histological approaches. We describe an IL-22-specific gene signature in the lung tissue of influenza A virus (IAV)-infected (and naive) mice that might explain the observed effects. Indeed, exogenous IL-22 modulates the gene expression profile in a way that suggests reinforcement of tissue integrity. Our results open the way to alternative approaches for limiting postinfluenza bacterial superinfection, particularly, systemic bacterial invasion.


Subject(s)
Interleukins/therapeutic use , Lung/immunology , Orthomyxoviridae Infections/drug therapy , Pneumococcal Infections/immunology , Superinfection/immunology , Animals , Humans , Immunotherapy , Liver/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Transcriptome , Interleukin-22
14.
J Immunol ; 197(8): 3225-3232, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27605012

ABSTRACT

Exogenous activation of invariant NKT (iNKT) cells by the superagonist α-galactosylceramide (α-GalCer) can protect against cancer, autoimmune diseases, and infections. In the current study, we investigated the effect of α-GalCer against Bacillus anthracis infection, the agent of anthrax. Using an experimental model of s.c. B. anthracis infection (an encapsulated nontoxigenic strain), we show that concomitant administration of α-GalCer delayed B. anthracis systemic dissemination and prolonged mouse survival. Depletion of subcapsular sinus CD169-positive macrophages by clodronate-containing liposome was associated with a lack of iNKT cell activation in the draining lymph nodes (dLNs) and prevented the protective effect of α-GalCer on bacterial dissemination out of the dLNs. Production of IFN-γ triggered chemokine (C-C motif) ligand 3 synthesis and recruitment of neutrophils in the dLNs, leading to the restraint of B. anthracis dissemination. Our data highlight a novel immunological pathway leading to the control of B. anthracis infection, a finding that might lead to improved therapeutics based on iNKT cells.


Subject(s)
Anthrax/immunology , Anthrax/microbiology , Bacillus anthracis/immunology , Natural Killer T-Cells/immunology , Animals , Anthrax/therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
PLoS Pathog ; 10(10): e1004300, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25299581

ABSTRACT

Non-conventional T lymphocytes constitute a special arm of the immune system and act as sentinels against pathogens at mucosal surfaces. These non-conventional T cells (including mucosal-associated invariant T [MAIT] cells, gamma delta [γδ] T cells, and natural killer T [NKT] cells) display several innate cell-like features and are rapidly activated by the recognition of conserved, stress-induced, self, and microbial ligands. Here, we review the role of non-conventional T cells during respiratory infections, with a particular focus on the encapsulated extracellular pathogen Streptococcus pneumoniae, the leading cause of bacterial pneumonia worldwide. We consider whether MAIT cells, γδ T cells, and NKT cells might offer opportunities for preventing and/or treating human pneumococcus infections.


Subject(s)
Natural Killer T-Cells/immunology , Pneumococcal Infections/immunology , Respiratory Tract Infections/immunology , Streptococcus pneumoniae/isolation & purification , T-Lymphocyte Subsets/immunology , Animals , Humans , Mucous Membrane/immunology , Respiratory Tract Infections/etiology , Streptococcus pneumoniae/immunology
17.
J Immunol ; 193(2): 961-9, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24913977

ABSTRACT

Immunotherapy aiming at enhancing innate and acquired host immunity is a promising approach for cancer treatment. The invariant NKT (iNKT) cell ligand α-galactosylceramide (α-GalCer) holds great promise in cancer therapy, although several concerns limit its use in clinics, including the uncontrolled response it promotes when delivered in a nonvectorized form. Therefore, development of delivery systems to in vivo target immune cells might be a valuable option to optimize iNKT cell-based antitumor responses. Using dendritic cell (DC)-depleted mice, DC transfer experiments, and in vivo active cell targeting, we show that presentation of α-GalCer by DCs not only triggers optimal primary iNKT cell stimulation, but also maintains secondary iNKT cell activation after challenge. Furthermore, targeted delivery of α-GalCer to CD8α(+) DCs, by means of anti-DEC205 decorated nanoparticles, enhances iNKT cell-based transactivation of NK cells, DCs, and γδ T cells. We report that codelivery of α-GalCer and protein Ag to CD8α(+) DCs triggers optimal Ag-specific Ab and cytotoxic CD8(+) T cell responses. Finally, we show that targeting nanoparticles containing α-GalCer and Ag to CD8α(+) DCs promotes potent antitumor responses, both in prophylactic and in therapeutic settings. Our data may have important implications in tumor immunotherapy and vaccine development.


Subject(s)
CD8 Antigens/immunology , Dendritic Cells/immunology , Galactosylceramides/immunology , Natural Killer T-Cells/immunology , Neoplasms, Experimental/immunology , Animals , Antibodies/chemistry , Antibodies/immunology , Antigen Presentation/immunology , Antigens, CD/immunology , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Dendritic Cells/metabolism , Drug Delivery Systems/methods , Galactosylceramides/administration & dosage , Galactosylceramides/chemistry , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Natural Killer T-Cells/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Cell Surface/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Burden/immunology
18.
Antimicrob Agents Chemother ; 59(10): 6064-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26195519

ABSTRACT

Prophylactic intranasal administration of the Toll-like receptor 5 (TLR5) agonist flagellin protects mice against respiratory pathogenic bacteria. We hypothesized that TLR5-mediated stimulation of lung immunity might improve the therapeutic index of antibiotics for the treatment of Streptococcus pneumoniae respiratory infections in mice. Intranasal administration of flagellin was combined with either oral administration of amoxicillin or intraperitoneal injection of trimethoprim-sulfamethoxazole to treat S. pneumoniae-infected animals. Compared with standalone treatments, the combination of antibiotic and flagellin resulted in a lower bacterial load in the lungs and greater protection against S. pneumoniae dissemination and was associated with an early increase in neutrophil infiltration in the airways. The antibiotic-flagellin combination treatment was, however, not associated with any exacerbation of inflammation. Moreover, combination treatment was more efficacious than standalone antibiotic treatments in the context of post-influenza virus pneumococcal infection. Lastly, TLR5 signaling was shown to be mandatory for the efficacy of the combined antibacterial therapy. This report is the first to show that combining antibiotic treatment with the stimulation of mucosal innate immunity is a potent antibacterial strategy against pneumonia.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Flagellin/therapeutic use , Pneumococcal Infections/drug therapy , Toll-Like Receptor 5/agonists , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Amoxicillin/therapeutic use , Animals , Female , Immunity, Innate/drug effects , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity
19.
J Immunol ; 191(5): 2344-50, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918982

ABSTRACT

Follicular helper T (TFH) cells are specialized in providing help for B cell differentiation and Ab secretion. Several positive and negative regulators of TFH cell differentiation have been described but their control is not fully understood. In this study, we show that Notch signaling in T cells is a major player in the development and function of TFH cells. T cell-specific gene ablation of Notch1 and Notch2 impaired differentiation of TFH cells in draining lymph nodes of mice immunized with T-dependent Ags or infected with parasites. Impaired TFH cell differentiation correlated with deficient germinal center development and the absence of high-affinity Abs. The impact of loss of Notch on TFH cell differentiation was largely independent of its effect on IL-4. These results show a previously unknown role for Notch in the regulation of TFH cell differentiation and function with implications for the control of this T cell population.


Subject(s)
Cell Differentiation/immunology , Lymphocyte Activation/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Flow Cytometry , Gene Knockdown Techniques , Germinal Center/cytology , Germinal Center/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Helper-Inducer/cytology
20.
J Infect Dis ; 210(3): 493-503, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24577508

ABSTRACT

Mucosal sites are continuously exposed to pathogenic microorganisms and are therefore equipped to control respiratory infections. Type 3 innate lymphoid cells (ILC3) are key players in antimicrobial defense in intestinal mucosa, through interleukin 17 and interleukin 22 (IL-22) production. The present study aimed at analyzing the distribution and function of ILC3 in the respiratory tract. We first observed that lung mucosa harbors a discrete population of ILC3 expressing CD127, CD90, CCR6, and the transcriptional factor RORγt. In addition, lung ILC3 were identified as a major source of IL-22 in response to interleukin 23 stimulation. During Streptococcus pneumoniae infection, ILC3 rapidly accumulated in the lung tissue to produce IL-22. In response to S. pneumoniae, dendritic cells and MyD88, an important adaptor of innate immunity, play critical functions in IL-22 production by ILC3. Finally, administration of the Toll-like receptor 5 agonist flagellin during S. pneumoniae challenge exacerbated IL-22 production by ILC3, a process that protects against lethal infection. In conclusion, boosting lung ILC3 might represent an interesting strategy to fight respiratory bacterial infections.


Subject(s)
Interleukins/metabolism , Lung/metabolism , Lymphocytes/classification , Lymphocytes/physiology , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Animals , Female , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukins/genetics , Lymphocyte Activation , Mice , Mice, Knockout , Streptococcus pneumoniae , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL