Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
J Immunol ; 208(7): 1675-1685, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35321877

RESUMEN

Strategically located at mucosal sites, mast cells are instrumental in sensing invading pathogens and modulating the quality of the ensuing immune responses depending on the nature of the infecting microbe. It is believed that mast cells produce type I IFN (IFN-I) in response to viruses, but not to bacterial infections, because of the incapacity of bacterial pathogens to internalize within mast cells, where signaling cascades leading to IFN-I production are generated. However, we have previously reported that, in contrast with other bacterial pathogens, Staphylococcus aureus can internalize into mast cells and therefore could trigger a unique response. In this study, we have investigated the molecular cross-talk between internalized S. aureus and the human mast cells HMC-1 using a dual RNA sequencing approach. We found that a proportion of internalized S. aureus underwent profound transcriptional reprogramming within HMC-1 cells to adapt to the nutrients and stress encountered in the intracellular environment and remained viable. HMC-1 cells, in turn, recognized intracellular S. aureus via cGMP-AMP synthase-STING-TANK-binding kinase 1 signaling pathway, leading to the production of IFN-I. Bacterial internalization and viability were crucial for IFN-I induction because inhibition of S. aureus internalization or infection with heat-killed bacteria completely prevented the production of IFN-I by HMC-1 cells. Feeding back in an autocrine manner in S. aureus-harboring HMC-1 cells and in a paracrine manner in noninfected neighboring HMC-1 cells, IFN-I promoted a cell-autonomous antimicrobial state by inducing the transcription of IFN-I-stimulated genes. This study provides unprecedented evidence of the capacity of mast cells to produce IFN-I in response to a bacterial pathogen.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Citosol , Humanos , Inmunidad Celular , Mastocitos
2.
Cell Mol Life Sci ; 80(8): 221, 2023 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-37480485

RESUMEN

Staphylococcus aureus is an important cause of chronic infections resulting from the failure of the host to eliminate the pathogen. Effective S. aureus clearance requires CD4+ T cell-mediated immunity. We previously showed that myeloid-derived suppressor cells (MDSC) expand during staphylococcal infections and support infection chronicity by inhibiting CD4+ T cell responses. The aim of this study was to elucidate the mechanisms underlying the suppressive effect exerted by MDSC on CD4+ T cells during chronic S. aureus infection. It is well known that activated CD4+ T cells undergo metabolic reprogramming from oxidative metabolism to aerobic glycolysis to meet their increased bioenergetic requirements. In this process, pyruvate is largely transformed into lactate by lactate dehydrogenase with the concomitant regeneration of NAD+, which is necessary for continued glycolysis. The by-product lactate needs to be excreted to maintain the glycolytic flux. Using SCENITH (single-cell energetic metabolism by profiling translation inhibition), we demonstrated here that MDSC inhibit CD4+ T cell responses by interfering with their metabolic activity. MDSC are highly glycolytic and excrete large amount of lactate in the local environment that alters the transmembrane concentration gradient and prevent removal of lactate by activated CD4+ T. Accumulation of endogenous lactate impedes the regeneration of NAD+, inhibit NAD-dependent glycolytic enzymes and stop glycolysis. Together, the results of this study have uncovered a role for metabolism on MDSC suppression of CD4+ T cell responses. Thus, reestablishment of their metabolic activity may represent a mean to improve the functionality of CD4+ T cells during chronic S. aureus infection.


Asunto(s)
Células Supresoras de Origen Mieloide , Infecciones Estafilocócicas , Humanos , Linfocitos T CD4-Positivos/metabolismo , Staphylococcus aureus/metabolismo , NAD/metabolismo , Infecciones Estafilocócicas/metabolismo , Lactatos/metabolismo
3.
J Allergy Clin Immunol ; 144(4S): S4-S18, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30468774

RESUMEN

Mast cells (MCs), which are well known for their effector functions in TH2-skewed allergic and also autoimmune inflammation, have become increasingly acknowledged for their role in protection of health. It is now clear that they are also key modulators of immune responses at interface organs, such as the skin or gut. MCs can prime tissues for adequate inflammatory responses and cooperate with dendritic cells in T-cell activation. They also regulate harmful immune responses in trauma and help to successfully orchestrate pregnancy. This review focuses on the beneficial effects of MCs on tissue homeostasis and elimination of toxins or venoms. MCs can enhance pathogen clearance in many bacterial, viral, and parasitic infections, such as through Toll-like receptor 2-triggered degranulation, secretion of antimicrobial cathelicidins, neutrophil recruitment, or provision of extracellular DNA traps. The role of MCs in tumors is more ambiguous; however, encouraging new findings show they can change the tumor microenvironment toward antitumor immunity when adequately triggered. Uterine tissue remodeling by α-chymase (mast cell protease [MCP] 5) is crucial for successful embryo implantation. MCP-4 and the tryptase MCP-6 emerge to be protective in central nervous system trauma by reducing inflammatory damage and excessive scar formation, thereby protecting axon growth. Last but not least, proteases, such as carboxypeptidase A, released by FcεRI-activated MCs detoxify an increasing number of venoms and endogenous toxins. A better understanding of the plasticity of MCs will help improve these advantageous effects and hint at ways to cut down detrimental MC actions.


Asunto(s)
Inmunidad Innata , Infecciones/inmunología , Mastocitos/inmunología , Animales , Catelicidinas/metabolismo , Degranulación de la Célula , Implantación del Embrión , Femenino , Homeostasis , Humanos , Embarazo , Receptor Toll-Like 2/metabolismo
4.
Int J Med Microbiol ; 308(6): 625-630, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28939437

RESUMEN

Staphylococcus aureus poses a significant public-health problem. Infection caused by S. aureus can manifest as acute or long-lasting persistent diseases that are often refractory to antibiotic and are associated with significant morbidity and mortality. To develop more effective strategies for preventing or treating these infections, it is crucial to understand why the immune response is incapable to eradicate the bacterium. When S. aureus first infect the host, there is a robust activation of the host innate immune responses. Generally, S. aureus can survive this initial interaction due to the expression of a wide array of virulence factors that interfere with the host innate immune defenses. After this initial interaction the acquired immune response is the arm of the host defenses that will try to clear the pathogen. However, S. aureus is capable of maintaining infection in the host even in the presence of a robust antigen-specific immune response. Thus, understanding the mechanisms underlying the ability of S. aureus to escape immune surveillance by the acquired immune response will help uncover potentially important targets for the development of immune-based adjunctive therapies and more efficient vaccines. There are several lines of evidence that lead us to believe that S. aureus can directly or indirectly disable the acquired immune response. This review will discuss the different immune evasion strategies used by S. aureus to modulate the different components of the acquired immune defenses.


Asunto(s)
Inmunidad Adaptativa , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Humanos , Inmunidad Humoral , Inmunidad Innata , Vigilancia Inmunológica , Ratones , Staphylococcus aureus/patogenicidad , Linfocitos T/inmunología , Factores de Virulencia
5.
J Infect Dis ; 216(11): 1444-1451, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29029332

RESUMEN

We have previously reported that myeloid-derived suppressor cells (MDSC), which are a heterogeneous population of immunosuppressive immature myeloid cells, expanded during chronic Staphylococcus aureus infection and promoted bacterial persistence by inhibiting effector T cells. Two major MDSC subsets, including monocytic MDSC and granulocytic MDSC, have been described to date. Here, we identified a new subset of MDSC (Eo-MDSC) in S. aureus-infected mice that phenotypically resembles eosinophils. Eo-MDSC exhibit eosinophilic cytoplasmic granules and express CD11b, the eosinophil marker Syglec-F, variable levels of CCR3, and low levels of interleukin-5Rα. Furthermore, Eo-MDSC accumulated at the site of infection and exerted a potent immunosuppressive effect on T-cell responses that was mediated by nitric oxide-dependent depletion of l-arginine. Increases in the number of Eo-MDSC by adoptive transfer caused a significant exacerbation of infection in S. aureus-infected mice. This study sheds new light on the heterogeneity and complexity of MDSC during chronic infection.


Asunto(s)
Eosinófilos/efectos de los fármacos , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/microbiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Traslado Adoptivo , Animales , Arginina , Antígeno CD11b/metabolismo , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Citocinas/biosíntesis , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Tolerancia Inmunológica/inmunología , Subunidad alfa del Receptor de Interleucina-5/metabolismo , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/patología , Óxido Nítrico , Fenotipo , Receptores CCR3/metabolismo , Bazo/microbiología , Bazo/patología , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/fisiología , Linfocitos T/efectos de los fármacos , Linfocitos T Reguladores
6.
Cell Microbiol ; 18(6): 807-19, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26595647

RESUMEN

Mast cells (MCs) are important sentinels of the host defence against invading pathogens. We previously reported that Staphylococcus aureus evaded the extracellular antimicrobial activities of MCs by promoting its internalization within these cells via ß1 integrins. Here, we investigated the molecular mechanisms governing this process. We found that S. aureus responded to the antimicrobial mediators released by MCs by up-regulating the expression of α-hemolysin (Hla), fibronectin-binding protein A and several regulatory systems. We also found that S. aureus induced the up-regulation of ß1 integrin expression on MCs and that this effect was mediated by Hla-ADAM10 (a disintegrin and metalloproteinase 10) interaction. Thus, deletion of Hla or inhibition of Hla-ADAM10 interaction significantly impaired S. aureus internalization within MCs. Furthermore, purified Hla but not the inactive HlaH35L induced up-regulation of ß1 integrin expression in MCs in a dose-dependent manner. Our data support a model in which S. aureus counter-reacts the extracellular microbicidal mechanisms of MCs by increasing expression of fibronectin-binding proteins and by inducing Hla-ADAM10-mediated up-regulation of ß1 integrin in MCs. The up-regulation of bacterial fibronectin-binding proteins, concomitantly with the increased expression of its receptor ß1 integrin on the MCs, resulted in enhanced S. aureus internalization through the binding of fibronectin-binding proteins to integrin ß1 via fibronectin.


Asunto(s)
Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Integrina beta1/metabolismo , Mastocitos/microbiología , Staphylococcus aureus/fisiología , Staphylococcus aureus/patogenicidad , Proteína ADAM10/metabolismo , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Toxinas Bacterianas/genética , Células Cultivadas , Femenino , Regulación Bacteriana de la Expresión Génica , Proteínas Hemolisinas/genética , Interacciones Huésped-Patógeno , Mastocitos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Infecciones Cutáneas Estafilocócicas/metabolismo , Infecciones Cutáneas Estafilocócicas/microbiología , Regulación hacia Arriba
7.
Proc Natl Acad Sci U S A ; 110(19): 7820-5, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610393

RESUMEN

Immunoresponsive gene 1 (Irg1) is highly expressed in mammalian macrophages during inflammation, but its biological function has not yet been elucidated. Here, we identify Irg1 as the gene coding for an enzyme producing itaconic acid (also known as methylenesuccinic acid) through the decarboxylation of cis-aconitate, a tricarboxylic acid cycle intermediate. Using a gain-and-loss-of-function approach in both mouse and human immune cells, we found Irg1 expression levels correlating with the amounts of itaconic acid, a metabolite previously proposed to have an antimicrobial effect. We purified IRG1 protein and identified its cis-aconitate decarboxylating activity in an enzymatic assay. Itaconic acid is an organic compound that inhibits isocitrate lyase, the key enzyme of the glyoxylate shunt, a pathway essential for bacterial growth under specific conditions. Here we show that itaconic acid inhibits the growth of bacteria expressing isocitrate lyase, such as Salmonella enterica and Mycobacterium tuberculosis. Furthermore, Irg1 gene silencing in macrophages resulted in significantly decreased intracellular itaconic acid levels as well as significantly reduced antimicrobial activity during bacterial infections. Taken together, our results demonstrate that IRG1 links cellular metabolism with immune defense by catalyzing itaconic acid production.


Asunto(s)
Regulación de la Expresión Génica , Hidroliasas/metabolismo , Macrófagos/metabolismo , Proteínas/metabolismo , Succinatos/metabolismo , Animales , Carboxiliasas , Catálisis , Línea Celular , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Inflamación , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Mycobacterium tuberculosis/metabolismo , ARN Interferente Pequeño/metabolismo
8.
Microbiology (Reading) ; 161(Pt 3): 621-627, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25533444

RESUMEN

Recent work has shown that coagulation and innate immunity are tightly interwoven host responses that help eradicate an invading pathogen. Some bacterial species, including Staphylococcus aureus, secrete pro-coagulant factors that, in turn, can modulate these immune reactions. Such mechanisms may not only protect the micro-organism from a lethal attack, but also promote bacterial proliferation and the establishment of infection. Our data showed that coagulase-positive S. aureus bacteria promoted clotting of plasma which was not seen when a coagulase-deficient mutant strain was used. Furthermore, in vitro studies showed that this ability constituted a mechanism that supported the aggregation, survival and persistence of the micro-organism within the fibrin network. These findings were also confirmed when agglutination and persistence of coagulase-positive S. aureus bacteria at the local focus of infection were studied in a subcutaneous murine infection model. In contrast, the coagulase-deficient S. aureus strain which was not able to induce clotting failed to aggregate and to persist in vivo. In conclusion, our data suggested that coagulase-positive S. aureus have evolved mechanisms that prevent their elimination within a fibrin clot.


Asunto(s)
Coagulación Sanguínea , Fibrina/metabolismo , Evasión Inmune , Infecciones Estafilocócicas/sangre , Staphylococcus aureus/inmunología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Coagulasa/genética , Coagulasa/metabolismo , Fibrina/genética , Humanos , Ratones , Ratones Endogámicos CBA , Viabilidad Microbiana , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/enzimología , Staphylococcus aureus/genética , Staphylococcus aureus/crecimiento & desarrollo
9.
Angew Chem Int Ed Engl ; 54(43): 12597-601, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26332072

RESUMEN

Isoniazid-filled Fe2 O3 hollow nanospheres (INH@Fe2 O3 , diameter <30 nm, 48 wt % INH-load) are prepared for the first time and suggested for tuberculosis therapy. After dextran-functionalization, the INH@Fe2 O3 @DEX nanocontainers show strong activity against Mycobacterium tuberculosis (M.tb.) and M.tb.-infected macrophages. The nanocontainers can be considered as "Trojan horses" and show efficient, active uptake into both M.tb.-infected macrophages and even into mycobacterial cells.


Asunto(s)
Antituberculosos/administración & dosificación , Antituberculosos/farmacología , Compuestos Férricos/química , Isoniazida/administración & dosificación , Isoniazida/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Nanosferas/química , Animales , Células Cultivadas , Humanos , Macrófagos/microbiología , Ratones , Nanosferas/ultraestructura , Tuberculosis/tratamiento farmacológico
10.
Infect Immun ; 82(11): 4496-507, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25114115

RESUMEN

Enterococcus faecalis has emerged as an important cause of life-threatening multidrug-resistant bacterial infections in the hospital setting. The pathogenesis of enterococcal infections has remained a relatively neglected field despite their obvious clinical relevance. The objective of this study was to characterize the interactions between mast cells (MCs), an innate immune cell population abundant in the intestinal lamina propria, and E. faecalis. This study was conducted with primary bone marrow-derived murine MCs. The results demonstrated that MCs exerted an antimicrobial effect against E. faecalis that was mediated both by degranulation, with the concomitant discharge of the antimicrobial effectors contained in the granules, and by the release of extracellular traps, in which E. faecalis was snared and killed. In particular, the cathelicidin LL-37 released by the MCs had potent antimicrobial effect against E. faecalis. We also investigated the specific receptors involved in the recognition of E. faecalis by MCs. We found that Toll-like receptors (TLRs) are critically involved in the MC recognition of E. faecalis, since MCs deficient in the expression of MyD88, an adaptor molecule required for signaling by most TLRs, were significantly impaired in their capacity to degranulate, to reduce E. faecalis growth as well as to release tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) after encountering this pathogen. Furthermore, TLR2 was identified as the most prominent TLR involved in the recognition of E. faecalis by MCs. The results of this study indicate that MCs may be important contributors to the host innate immune defenses against E. faecalis.


Asunto(s)
Enterococcus faecalis/fisiología , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/microbiología , Mastocitos/fisiología , Animales , Adhesión Bacteriana , Células de la Médula Ósea/fisiología , Células Cultivadas , Enterococcus faecalis/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Organismos Libres de Patógenos Específicos , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo
11.
Front Immunol ; 15: 1328193, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38380317

RESUMEN

Regulatory cells, such as regulatory T cells (Tregs), regulatory B cells (Bregs), and myeloid-derived suppressor cells (MDSCs), play a crucial role in preserving immune tolerance and controlling immune responses during infections to prevent excessive immune activation. However, pathogens have developed strategies to hijack these regulatory cells to decrease the overall effectiveness of the immune response and persist within the host. Consequently, therapeutic targeting of these immunosuppressive mechanisms during infection can reinvigorate the immune response and improve the infection outcome. The suppressive mechanisms of regulatory cells are not only numerous but also redundant, reflecting the complexity of the regulatory network in modulating the immune responses. The context of the immune response, such as the type of pathogen or tissue involved, further influences the regulatory mechanisms involved. Examples of these immunosuppressive mechanisms include the production of inhibitory cytokines such as interleukin 10 (IL-10) and transforming growth factor beta (TGF-ß) that inhibit the production of pro-inflammatory cytokines and dampen the activation and proliferation of effector T cells. In addition, regulatory cells utilize inhibitory receptors like cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) to engage with their respective effector cells, thereby suppressing their function. An alternative approach involves the modulation of metabolic reprogramming in effector immune cells to limit their activation and proliferation. In this review, we provide an overview of the major mechanisms mediating the immunosuppressive effect of the different regulatory cell subsets in the context of infection.


Asunto(s)
Tolerancia Inmunológica , Linfocitos T Reguladores , Citocinas , Factor de Crecimiento Transformador beta , Terapia de Inmunosupresión
12.
Am J Pathol ; 181(4): 1327-37, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22885107

RESUMEN

Dendritic cells (DCs) play an important role in integration of the immune responses induced by pathogens. The purpose of this study was to determine the importance of DCs in host defense against Staphylococcus aureus bacteremia. Using a murine infection model, we demonstrated that DCs are rapidly recruited into infected tissue after intravenous inoculation with S. aureus. The recruited DCs were fully functional and in a more advanced stage of maturation than those isolated from uninfected mice. Depletion of DCs in CD11c-DTR transgenic mice resulted in substantial worsening of infection, as indicated by increased bacterial loads in kidneys and lungs, accelerated mortality, and more severe pathology. Furthermore, DC depletion completely abolished IL-12 production in response to infection. The beneficial effect afforded by DCs during S. aureus infection was not mediated by their contribution to direct bacterial killing, nor by increased neutrophil recruitment. Instead, neutrophil influx (along with expression of CXC chemokines) was significantly enhanced in infected tissue after depletion of DCs. We also found that the bactericidal capacity of the recruited neutrophils was significantly impaired in DC-depleted mice. More importantly, the detrimental effect of DC depletion was practically reversed by treatment with exogenous recombinant mouse IL-12. Our results demonstrated that DCs, probably through their production of IL-12, play an important role in coordinating the inflammatory response during S. aureus infection.


Asunto(s)
Células Dendríticas/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad/inmunología , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Bacteriemia/sangre , Bacteriemia/complicaciones , Bacteriemia/microbiología , Bacteriemia/patología , Antígeno CD11c/metabolismo , Células Dendríticas/microbiología , Células Dendríticas/patología , Células Dendríticas/ultraestructura , Interacciones Huésped-Patógeno/efectos de los fármacos , Inmunidad/efectos de los fármacos , Inflamación/sangre , Inflamación/complicaciones , Inflamación/patología , Interleucina-12/administración & dosificación , Interleucina-12/metabolismo , Interleucina-12/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Viabilidad Microbiana/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/microbiología , Bazo/patología , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/ultraestructura
13.
Front Microbiol ; 14: 1228472, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37965557

RESUMEN

Streptococcus canis is a zoonotic agent that causes severe invasive diseases in domestic animals and humans, but little is known about its pathogenesis and virulence mechanisms so far. SCM, the M-like protein expressed by S. canis, is considered one of the major virulence determinants. Here, we report on the two distinct groups of SCM. SCM-1 proteins were already described to interact with its ligands IgG and plasminogen as well as with itself and confer antiphagocytic capability of SCM-1 expressing bacterial isolates. In contrast, the function of SCM-2 type remained unclear to date. Using whole-genome sequencing and subsequent bioinformatics, FACS analysis, fluorescence microscopy and surface plasmon resonance spectrometry, we demonstrate that, although different in amino acid sequence, a selection of diverse SCM-2-type S. canis isolates, phylogenetically representing the full breadth of SCM-2 sequences, were able to bind fibrinogen. Using targeted mutagenesis of an SCM-2 isolate, we further demonstrated that this strain was significantly less able to survive in canine blood. With respect to similar studies showing a correlation between fibrinogen binding and survival in whole blood, we hypothesize that SCM-2 has an important contribution to the pathogenesis of S. canis in the host.

14.
J Immunol ; 185(4): 2372-81, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20644176

RESUMEN

Streptococcus pyogenes is a significant human pathogen that can cause life-threatening invasive infections. Understanding the mechanism of disease is crucial to the development of more effective therapies. In this report, we explored the role of PGE(2), an arachidonic acid metabolite, and its rate-limiting enzyme cyclooxygenase 2 (COX-2) in the pathogenesis of severe S. pyogenes infections. We found that the COX-2 expression levels in tissue biopsies from S. pyogenes-infected patients, as well as in tissue of experimentally infected mice, strongly correlated with the severity of infection. This harmful effect was attributed to PGE(2)-mediated suppression of the bactericidial activity of macrophages through interaction with the G2-coupled E prostanoid receptor. The suppressive effect of PGE(2) was associated with enhanced intracellular cAMP production and was mimicked by the cAMP-elevating agent, forskolin. Activation of protein kinase A (PKA) was the downstream effector mechanisms of cAMP because treatment with PKI(14-22), a highly specific inhibitor of PKA, prevented the PGE(2)-mediated inhibition of S. pyogenes killing in macrophages. The inhibitory effect exerted by PKA in the generation of antimicrobial oxygen radical species seems to be the ultimate effector mechanism responsible for the PGE(2)-mediated downregulation of the macrophage bactericidal activity. Importantly, either genetic ablation of COX-2, pharmacological inhibition of COX-2 or treatment with the G2-coupled E prostanoid antagonist, AH6809, significantly improved the disease outcome in S. pyogenes infected mice. Therefore, the results of this study open up new perspectives on potential molecular pathways that are prone to pharmacological manipulation during severe streptococcal infections.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Macrófagos/metabolismo , Infecciones Estreptocócicas/metabolismo , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa/farmacología , Dinoprostona/antagonistas & inhibidores , Femenino , Interacciones Huésped-Patógeno , Humanos , Inmunohistoquímica , Macrófagos/citología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Nitrobencenos/farmacología , Antagonistas de Prostaglandina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Índice de Severidad de la Enfermedad , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/fisiología , Sulfonamidas/farmacología , Xantonas/farmacología
15.
J Innate Immun ; 14(3): 257-274, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34763332

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a compendium of immature myeloid cells that exhibit potent T-cell suppressive capacity and expand during pathological conditions such as cancer and chronic infections. Although well-characterized in cancer, the physiology of MDSCs in the infection setting remains enigmatic. Here, we integrated single-cell RNA sequencing (scRNA-seq) and functional metabolic profiling to gain deeper insights into the factors governing the generation and maintenance of MDSCs in chronic Staphylococcus aureus infection. We found that MDSCs originate not only in the bone marrow but also at extramedullary sites in S. aureus-infected mice. scRNA-seq showed that infection-driven MDSCs encompass a spectrum of myeloid precursors in different stages of differentiation, ranging from promyelocytes to mature neutrophils. Furthermore, the scRNA-seq analysis has also uncovered valuable phenotypic markers to distinguish mature myeloid cells from immature MDSCs. Metabolic profiling indicates that MDSCs exhibit high glycolytic activity and high glucose consumption rates, which are required for undergoing terminal maturation. However, rapid glucose consumption by MDSCs added to infection-induced perturbations in the glucose supplies in infected mice hinders the terminal maturation of MDSCs and promotes their accumulation in an immature stage. In a proof-of-concept in vivo experiment, we demonstrate the beneficial effect of increasing glucose availability in promoting MDSC terminal differentiation in infected mice. Our results provide valuable information of how metabolic alterations induced by infection influence reprogramming and differentiation of MDSCs.


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias , Infecciones Estafilocócicas , Animales , Glucosa , Ratones , Células Supresoras de Origen Mieloide/fisiología , Infección Persistente , Staphylococcus aureus
16.
mBio ; 13(5): e0231622, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36102512

RESUMEN

Staphylococcus aureus is a leading cause of difficult-to-treat infections. The capacity of S. aureus to survive and persist within phagocytic cells is an important factor contributing to therapy failures and infection recurrence. Therefore, interfering with S. aureus intracellular persistence is key to treatment success. In this study, we used a S. aureus strain carrying the reporter mKikumeGR that enables the monitoring of the metabolic status of intracellular bacteria to achieve a better understanding of the molecular mechanisms facilitating S. aureus survival and persistence within macrophages. We found that shortly after bacteria internalization, a large fraction of macrophages harbored mainly S. aureus with high metabolic activity. This population decreased gradually over time with the concomitant increase of a macrophage subpopulation harboring S. aureus with low metabolic activity, which prevailed at later times. A dual RNA-seq analysis performed in each macrophage subpopulation showed that the host transcriptional response was similar between both subpopulations. However, intracellular S. aureus exhibited disparate gene expression profiles depending on its metabolic state. Whereas S. aureus with high metabolic activity exhibited a greater expression of genes involved in protein synthesis and proliferation, bacteria with low metabolic activity displayed a higher expression of oxidative stress response-related genes, silenced genes involved in energy-consuming processes, and exhibited a dormant-like state. Consequently, we propose that reducing metabolic activity and entering into a dormant-like state constitute a survival strategy used by S. aureus to overcome the adverse environment encountered within macrophages and to persist in the intracellular niche. IMPORTANCE The capacity of Staphylococcus aureus to survive and persist within phagocytic cells has been associated with antibiotic treatment failure and recurrent infections. Here, we investigated the molecular mechanisms leading to S. aureus persistence within macrophages using a reporter system that enables to distinguish between intracellular bacteria with high and low metabolic activity in combinstion with a dual RNA-seq approach. We found that with the progression of infection, intracellular S. aureus transitions from a high metabolic state to a low metabolic dormant-like state by turning off major energy-consuming processes while remaining viable. This process seems to be driven by the level of stress encountered in the intracellular niche. Our study indicates that effective therapies by which to treat S. aureus infections should be able to target not only high metabolic bacteria but also intracellular dormant-like S. aureus.


Asunto(s)
Fenómenos Bioquímicos , Infecciones Estafilocócicas , Humanos , Staphylococcus aureus/genética , Infecciones Estafilocócicas/microbiología , Macrófagos/microbiología , Antibacterianos
17.
Biochim Biophys Acta Mol Basis Dis ; 1868(12): 166530, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36038039

RESUMEN

Macrophages undergo extensive metabolic reprogramming during classical pro-inflammatory polarization (M1-like). The accumulation of itaconate has been recognized as both a consequence and mediator of the inflammatory response. In this study we first examined the specific functions of itaconate inside fractionated mitochondria. We show that M1 macrophages produce itaconate de novo via aconitase decarboxylase 1 (ACOD1) inside mitochondria. The carbon for this reaction is not only supplied by oxidative TCA cycling, but also through the reductive carboxylation of α-ketoglutarate by isocitrate dehydrogenase (IDH). While macrophages are capable of sustaining a certain degree of itaconate production during hypoxia by augmenting the activity of IDH-dependent reductive carboxylation, we demonstrate that sufficient itaconate synthesis requires a balance of reductive and oxidative TCA cycle metabolism in mouse macrophages. In comparison, human macrophages increase itaconate accumulation under hypoxic conditions by augmenting reductive carboxylation activity. We further demonstrated that itaconate attenuates reductive carboxylation at IDH2, restricting its own production and the accumulation of the immunomodulatory metabolites citrate and 2-hydroxyglutarate. In line with this, reductive carboxylation is enhanced in ACOD1-depleted macrophages. Mechanistically, the inhibition of IDH2 by itaconate is linked to the alteration of the mitochondrial NADP+/NADPH ratio and competitive succinate dehydrogenase inhibition. Taken together, our findings extend the current model of TCA cycle reprogramming during pro-inflammatory macrophage activation and identified novel regulatory properties of itaconate.


Asunto(s)
Carboxiliasas , Ciclo del Ácido Cítrico , Isocitrato Deshidrogenasa , Succinatos , Aconitato Hidratasa/metabolismo , Animales , Carbono/metabolismo , Carboxiliasas/metabolismo , Citratos , Retroalimentación , Humanos , Ácidos Cetoglutáricos/metabolismo , Ratones , NADP/metabolismo , Succinato Deshidrogenasa/metabolismo , Succinatos/metabolismo
18.
Nat Metab ; 4(5): 524-533, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35655024

RESUMEN

Since its discovery in inflammatory macrophages, itaconate has attracted much attention due to its antimicrobial and immunomodulatory activity1-3. However, instead of investigating itaconate itself, most studies used derivatized forms of itaconate and thus the role of non-derivatized itaconate needs to be scrutinized. Mesaconate, a metabolite structurally very close to itaconate, has never been implicated in mammalian cells. Here we show that mesaconate is synthesized in inflammatory macrophages from itaconate. We find that both, non-derivatized itaconate and mesaconate dampen the glycolytic activity to a similar extent, whereas only itaconate is able to repress tricarboxylic acid cycle activity and cellular respiration. In contrast to itaconate, mesaconate does not inhibit succinate dehydrogenase. Despite their distinct impact on metabolism, both metabolites exert similar immunomodulatory effects in pro-inflammatory macrophages, specifically a reduction of interleukin (IL)-6 and IL-12 secretion and an increase of CXCL10 production in a manner that is independent of NRF2 and ATF3. We show that a treatment with neither mesaconate nor itaconate impairs IL-1ß secretion and inflammasome activation. In summary, our results identify mesaconate as an immunomodulatory metabolite in macrophages, which interferes to a lesser extent with cellular metabolism than itaconate.


Asunto(s)
Macrófagos , Succinatos , Animales , Inflamasomas , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Succinatos/metabolismo , Succinatos/farmacología
19.
Am J Pathol ; 176(2): 754-63, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20019195

RESUMEN

Several in vitro studies have emphasized the importance of toll-like receptor/myeloid differentiation factor 88 (MyD88) signaling in the inflammatory response to Streptococcus pyogenes. Since the extent of inflammation has been implicated in the severity of streptococcal diseases, we have examined here the role of toll-like receptor/MyD88 signaling in the pathophysiology of experimental S. pyogenes infection. To this end, we compared the response of MyD88-knockout (MyD88(-/-)) after subcutaneous inoculation with S. pyogenes with that of C57BL/6 mice. Our results show that MyD88(-/-) mice harbored significantly more bacteria in the organs and succumbed to infection much earlier than C57BL/6 animals. Absence of MyD88 resulted in diminished production of inflammatory cytokines such as interleukin-12, interferon-gamma, and tumor necrosis factor-alpha as well as chemoattractants such as monocyte chemotactic protein-1 (MCP-1) and Keratinocyte-derived chemokine (KC), and hampered recruitment of effector cells involved in bacterial clearance (macrophages and neutrophils) to the infection site. Furthermore, MyD88(-/-) but not C57BL/6 mice exhibited a massive infiltration of eosinophils in infected organs, which can be explained by an impaired production of the regulatory chemokines, gamma interferon-induced monokine (MIG/CXCL9) and interferon-induced protein 10 (IP-10/CXCL10), which can inhibit transmigration of eosinophils. Our results indicate that MyD88 signaling targets effector cells to the site of streptococcal infection and prevents extravasation of cells that can induce tissue damage. Therefore, MyD88 signaling may be important for shaping the quality of the inflammatory response elicited during infection to ensure optimal effector functions.


Asunto(s)
Inflamación/genética , Factor 88 de Diferenciación Mieloide/genética , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Células Cultivadas , Quimiotaxis de Leucocito/genética , Citocinas/sangre , Predisposición Genética a la Enfermedad , Inflamación/sangre , Inflamación/inmunología , Inflamación/microbiología , Mediadores de Inflamación/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , Neutrófilos/inmunología , Fagocitosis/genética , Fagocitosis/inmunología , Infecciones Estreptocócicas/sangre , Streptococcus pyogenes/fisiología
20.
J Pathol ; 220(5): 521-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20020512

RESUMEN

Epidemiological studies have shown that the elderly are at higher risk of severe Streptococcus pyogenes infections. In this study, we used a mouse model that displays the age-related loss of resistance to S. pyogenes infection seen in humans to investigate the impaired immune mechanism underlying the age-associated susceptibility to this pathogen. Young (2-3 months old) and aged (>20 months old) BALB/c mice were subcutaneously or intravenously inoculated with S. pyogenes and their capacity to control infection was compared. Aged mice showed faster progression of disease, earlier morbidity, and increased mortality when compared with young animals. Since macrophages are critical for host defence against S. pyogenes, we investigated whether susceptibility of aged mice may be due to an age-associated decline in the functionality of these cells. Our results showed that macrophages from aged mice were as capable as those from young animals to uptake and kill S. pyogenes, but the number of resident tissue macrophages was significantly reduced in the aged host. Treatment of aged mice with macrophage colony-stimulating factor (M-CSF) significantly increased the number of resident macrophages and improved their response to infection. Our results indicate that treatment with M-CSF can restore, at least in part, the mechanisms affected by immunosenescence and enhance the natural resistance of aged mice to infection with S. pyogenes.


Asunto(s)
Envejecimiento/inmunología , Tolerancia Inmunológica/fisiología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/patogenicidad , Animales , Recuento de Células , Células Cultivadas , Citocinas/sangre , Susceptibilidad a Enfermedades , Femenino , Mediadores de Inflamación/sangre , Dosificación Letal Mediana , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/prevención & control , Streptococcus pyogenes/crecimiento & desarrollo , Análisis de Supervivencia , Virulencia/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA