Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 600
Filtrer
1.
Microbiol Spectr ; 10(1): e0254621, 2022 02 23.
Article de Anglais | MEDLINE | ID: mdl-35080463

RÉSUMÉ

Mycobacterium abscessus is the etiological agent of severe pulmonary infections in vulnerable patients, such as those with cystic fibrosis (CF), where it represents a relevant cause of morbidity and mortality. Treatment of pulmonary infections caused by M. abscessus remains extremely difficult, as this species is resistant to most classes of antibiotics, including macrolides, aminoglycosides, rifamycins, tetracyclines, and ß-lactams. Here, we show that apoptotic body like liposomes loaded with phosphatidylinositol 5-phosphate (ABL/PI5P) enhance the antimycobacterial response, both in macrophages from healthy donors exposed to pharmacological inhibition of cystic fibrosis transmembrane conductance regulator (CFTR) and in macrophages from CF patients, by enhancing phagosome acidification and reactive oxygen species (ROS) production. The treatment with liposomes of wild-type as well as CF mice, intratracheally infected with M. abscessus, resulted in about a 2-log reduction of pulmonary mycobacterial burden and a significant reduction of macrophages and neutrophils in bronchoalveolar lavage fluid (BALF). Finally, the combination treatment with ABL/PI5P and amikacin, to specifically target intracellular and extracellular bacilli, resulted in a further significant reduction of both pulmonary mycobacterial burden and inflammatory response in comparison with the single treatments. These results offer the conceptual basis for a novel therapeutic regimen based on antibiotic and bioactive liposomes, used as a combined host- and pathogen-directed therapeutic strategy, aimed at the control of M. abscessus infection, and of related immunopathogenic responses, for which therapeutic options are still limited. IMPORTANCE Mycobacterium abscessus is an opportunistic pathogen intrinsically resistant to many antibiotics, frequently linked to chronic pulmonary infections, and representing a relevant cause of morbidity and mortality, especially in immunocompromised patients, such as those affected by cystic fibrosis. M. abscessus-caused pulmonary infection treatment is extremely difficult due to its high toxicity and long-lasting regimen with life-impairing side effects and the scarce availability of new antibiotics approved for human use. In this context, there is an urgent need for the development of an alternative therapeutic strategy that aims at improving the current management of patients affected by chronic M. abscessus infections. Our data support the therapeutic value of a combined host- and pathogen-directed therapy as a promising approach, as an alternative to single treatments, to simultaneously target intracellular and extracellular pathogens and improve the clinical management of patients infected with multidrug-resistant pathogens such as M. abscessus.


Sujet(s)
Antibactériens/administration et posologie , Mucoviscidose/immunologie , Infections à mycobactéries non tuberculeuses/traitement médicamenteux , Mycobacterium abscessus/effets des médicaments et des substances chimiques , Phosphates phosphatidylinositol/administration et posologie , Amikacine/administration et posologie , Amikacine/composition chimique , Animaux , Antibactériens/composition chimique , Mucoviscidose/complications , Mucoviscidose/génétique , Mucoviscidose/microbiologie , Protéine CFTR/génétique , Protéine CFTR/immunologie , Femelle , Humains , Liposomes/composition chimique , Macrophages/immunologie , Mâle , Souris , Souris de lignée C57BL , Infections à mycobactéries non tuberculeuses/étiologie , Infections à mycobactéries non tuberculeuses/immunologie , Infections à mycobactéries non tuberculeuses/microbiologie , Mycobacterium abscessus/physiologie , Phagosomes/immunologie , Phosphates phosphatidylinositol/composition chimique , Espèces réactives de l'oxygène/immunologie
2.
Front Immunol ; 12: 687044, 2021.
Article de Anglais | MEDLINE | ID: mdl-34630380

RÉSUMÉ

Phagosome-lysosome fusion in innate immune cells like macrophages and neutrophils marshal an essential role in eliminating intracellular microorganisms. In microbe-challenged macrophages, phagosome-lysosome fusion occurs 4 to 6 h after the phagocytic uptake of the microbe. However, live pathogenic mycobacteria hinder the transfer of phagosomes to lysosomes, up to 20 h post-phagocytic uptake. This period is required to evade pro-inflammatory response and upregulate the acid-stress tolerant proteins. The exact sequence of events through which mycobacteria retards phagolysosome formation remains an enigma. The macrophage coat protein Coronin1(Cor1) is recruited and retained by mycobacteria on the phagosome membrane to retard its maturation by hindering the access of phagosome maturation factors. Mycobacteria-infected macrophages exhibit an increased cAMP level, and based on receptor stimulus, Cor1 expressing cells show a higher level of cAMP than non-Cor1 expressing cells. Here we have shown that infection of bone marrow-derived macrophages with H37Rv causes a Cor1 dependent rise of intracellular cAMP levels at the vicinity of the phagosomes. This increased cAMP fuels cytoskeletal protein Cofilin1 to depolymerize F-actin around the mycobacteria-containing phagosome. Owing to reduced F-actin levels, the movement of the phagosome toward the lysosomes is hindered, thus contributing to the retarded phagosome maturation process. Additionally, Cor1 mediated upregulation of Cofilin1 also contributes to the prevention of phagosomal acidification, which further aids in the retardation of phagosome maturation. Overall, our study provides first-hand information on Cor1 mediated retardation of phagosome maturation, which can be utilized in developing novel peptidomimetics as part of host-directed therapeutics against tuberculosis.


Sujet(s)
Cofiline-1/métabolisme , AMP cyclique/métabolisme , Macrophages/microbiologie , Protéines des microfilaments/métabolisme , Infections à mycobactéries non tuberculeuses/microbiologie , Mycobacterium bovis/pathogénicité , Mycobacterium smegmatis/pathogénicité , Mycobacterium tuberculosis/pathogénicité , Phagosomes/microbiologie , Tuberculose/microbiologie , Animaux , Lignée cellulaire , Interactions hôte-pathogène , Concentration en ions d'hydrogène , Macrophages/immunologie , Macrophages/métabolisme , Souris , Protéines des microfilaments/génétique , Infections à mycobactéries non tuberculeuses/immunologie , Infections à mycobactéries non tuberculeuses/métabolisme , Mycobacterium bovis/immunologie , Mycobacterium smegmatis/immunologie , Mycobacterium tuberculosis/immunologie , Phagosomes/immunologie , Phagosomes/métabolisme , Systèmes de seconds messagers , Tuberculose/immunologie , Tuberculose/métabolisme
3.
Front Immunol ; 12: 728848, 2021.
Article de Anglais | MEDLINE | ID: mdl-34557194

RÉSUMÉ

Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major, Leishmania braziliensis and Leishmania infantum. In general, the CD4+ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis, as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.


Sujet(s)
Immunité acquise , Système immunitaire/parasitologie , Immunité innée , Leishmania brasiliensis/pathogénicité , Leishmaniose cutanée/parasitologie , Leishmaniose viscérale/parasitologie , Phagocytose , Phagosomes/parasitologie , Animaux , Maladie chronique , Interactions hôte-parasite , Humains , Système immunitaire/immunologie , Système immunitaire/métabolisme , Leishmania brasiliensis/immunologie , Leishmaniose cutanée/immunologie , Leishmaniose cutanée/métabolisme , Leishmaniose cutanéomuqueuse/immunologie , Leishmaniose cutanéomuqueuse/métabolisme , Leishmaniose cutanéomuqueuse/parasitologie , Leishmaniose viscérale/immunologie , Leishmaniose viscérale/métabolisme , Phagosomes/immunologie , Phagosomes/métabolisme
4.
Nat Commun ; 12(1): 4999, 2021 08 17.
Article de Anglais | MEDLINE | ID: mdl-34404769

RÉSUMÉ

The type I interferon (IFN) signaling pathway has important functions in resistance to viral infection, with the downstream induction of interferon stimulated genes (ISG) protecting the host from virus entry, replication and spread. Listeria monocytogenes (Lm), a facultative intracellular foodborne pathogen, can exploit the type I IFN response as part of their pathogenic strategy, but the molecular mechanisms involved remain unclear. Here we show that type I IFN suppresses the antibacterial activity of phagocytes to promote systemic Lm infection. Mechanistically, type I IFN suppresses phagosome maturation and proteolysis of Lm virulence factors ActA and LLO, thereby promoting phagosome escape and cell-to-cell spread; the antiviral protein, IFN-induced transmembrane protein 3 (IFITM3), is required for this type I IFN-mediated alteration. Ifitm3-/- mice are resistant to systemic infection by Lm, displaying decreased bacterial spread in tissues, and increased immune cell recruitment and pro-inflammatory cytokine signaling. Together, our findings show how an antiviral mechanism in phagocytes can be exploited by bacterial pathogens, and implicate IFITM3 as a potential antimicrobial therapeutic target.


Sujet(s)
Antibactériens/pharmacologie , Listeria/effets des médicaments et des substances chimiques , Infections à Listeria/immunologie , Protéines membranaires/antagonistes et inhibiteurs , Protéines membranaires/métabolisme , Phagocytes/immunologie , Phagocytes/microbiologie , Animaux , Modèles animaux de maladie humaine , Interactions hôte-pathogène , Interféron de type I/métabolisme , Listeria monocytogenes/immunologie , Mâle , Protéines membranaires/génétique , Souris , Souris de lignée C57BL , Souris knockout , Phagosomes/immunologie , Cellules RAW 264.7 , Transcriptome , Facteurs de virulence , Pénétration virale/effets des médicaments et des substances chimiques
5.
mBio ; 12(4): e0124721, 2021 08 31.
Article de Anglais | MEDLINE | ID: mdl-34311577

RÉSUMÉ

Monocytes play an important role in the host defense against Plasmodium vivax as the main source of inflammatory cytokines and mitochondrial reactive oxygen species (mROS). Here, we show that monocyte metabolism is altered during human P. vivax malaria, with mitochondria playing a major function in this switch. The process involves a reprograming in which the cells increase glucose uptake and produce ATP via glycolysis instead of oxidative phosphorylation. P. vivax infection results in dysregulated mitochondrial gene expression and in altered membrane potential leading to mROS increase rather than ATP production. When monocytes were incubated with P. vivax-infected reticulocytes, mitochondria colocalized with phagolysosomes containing parasites representing an important source mROS. Importantly, the mitochondrial enzyme superoxide dismutase 2 (SOD2) is simultaneously induced in monocytes from malaria patients. Taken together, the monocyte metabolic reprograming with an increased mROS production may contribute to protective responses against P. vivax while triggering immunomodulatory mechanisms to circumvent tissue damage. IMPORTANCE Plasmodium vivax is the most widely distributed causative agent of human malaria. To achieve parasite control, the human immune system develops a substantial inflammatory response that is also responsible for the symptoms of the disease. Among the cells involved in this response, monocytes play an important role. Here, we show that monocyte metabolism is altered during malaria, with its mitochondria playing a major function in this switch. This change involves a reprograming process in which the cells increase glucose uptake and produce ATP via glycolysis instead of oxidative phosphorylation. The resulting altered mitochondrial membrane potential leads to an increase in mitochondrial reactive oxygen species rather than ATP. These data suggest that agents that change metabolism should be investigated and used with caution during malaria.


Sujet(s)
Mitochondries/métabolisme , Mitochondries/anatomopathologie , Monocytes/métabolisme , Monocytes/anatomopathologie , Plasmodium vivax/immunologie , Réticulocytes/parasitologie , Adénosine triphosphate/métabolisme , Adolescent , Adulte , Sujet âgé , Femelle , Expression des gènes , Glycolyse , Humains , Paludisme à Plasmodium vivax/immunologie , Paludisme à Plasmodium vivax/physiopathologie , Mâle , Adulte d'âge moyen , Mitochondries/génétique , Monocytes/cytologie , Monocytes/immunologie , Phagosomes/immunologie , Phagosomes/parasitologie , Plasmodium vivax/génétique , Plasmodium vivax/pathogénicité , Espèces réactives de l'oxygène/métabolisme , Superoxide dismutase/génétique , Superoxide dismutase/métabolisme , Jeune adulte
6.
mBio ; 12(3): e0100821, 2021 06 29.
Article de Anglais | MEDLINE | ID: mdl-34076467

RÉSUMÉ

The Dot/Icm type IV secretion system (T4SS) of Legionella pneumophila is essential for lysosomal evasion and permissiveness of macrophages for intracellular proliferation of the pathogen. In contrast, we show that polymorphonuclear cells (PMNs) respond to a functional Dot/Icm system through rapid restriction of L. pneumophila. Specifically, we show that the L. pneumophila T4SS-injected amylase (LamA) effector catalyzes rapid glycogen degradation in the PMNs cytosol, leading to cytosolic hyperglucose. Neutrophils respond through immunometabolic reprogramming that includes upregulated aerobic glycolysis. The PMNs become activated with spatial generation of intracellular reactive oxygen species within the Legionella-containing phagosome (LCP) and fusion of specific and azurophilic granules to the LCP, leading to rapid restriction of L. pneumophila. We conclude that in contrast to macrophages, PMNs respond to a functional Dot/Icm system, and specifically to the effect of the injected amylase effector, through rapid engagement of major microbicidal processes and rapid restriction of the pathogen. IMPORTANCE Legionella pneumophila is commonly found in aquatic environments and resides within a wide variety of amoebal hosts. Upon aerosol transmission to humans, L. pneumophila invades and replicates with alveolar macrophages, causing pneumonia designated Legionnaires' disease. In addition to alveolar macrophages, neutrophils infiltrate into the lungs of infected patients. Unlike alveolar macrophages, neutrophils restrict and kill L. pneumophila, but the mechanisms were previously unclear. Here, we show that the pathogen secretes an amylase (LamA) enzyme that rapidly breakdowns glycogen stores within neutrophils, and this triggers increased glycolysis. Subsequently, the two major killing mechanisms of neutrophils, granule fusion and production of reactive oxygen species, are activated, resulting in rapid killing of L. pneumophila.


Sujet(s)
Legionella pneumophila/immunologie , Granulocytes neutrophiles/microbiologie , Systèmes de sécrétion de type IV/immunologie , Protéines bactériennes/métabolisme , Cytosol/microbiologie , Glycogène/métabolisme , Glycolyse , Humains , Legionella pneumophila/génétique , Legionella pneumophila/métabolisme , Maladie des légionnaires/microbiologie , Phagosomes/immunologie , Phagosomes/microbiologie , Espèces réactives de l'oxygène/immunologie , Systèmes de sécrétion de type IV/génétique
7.
Sci Rep ; 11(1): 13430, 2021 06 28.
Article de Anglais | MEDLINE | ID: mdl-34183758

RÉSUMÉ

Many innate immune receptors function collaboratively to detect and elicit immune responses to pathogens, but the physical mechanisms that govern the interaction and signaling crosstalk between the receptors are unclear. In this study, we report that the signaling crosstalk between Fc gamma receptor (FcγR) and Toll-like receptor (TLR)2/1 can be overall synergistic or inhibitory depending on the spatial proximity between the receptor pair on phagosome membranes. Using a geometric manipulation strategy, we physically altered the spatial distribution of FcγR and TLR2 on single phagosomes. We demonstrate that the signaling synergy between FcγR and TLR2/1 depends on the proximity of the receptors and decreases as spatial separation between them increases. However, the inhibitory effect from FcγRIIb on TLR2-dependent signaling is always present and independent of receptor proximity. The overall cell responses are an integration from these two mechanisms. This study presents quantitative evidence that the nanoscale proximity between FcγR and TLR2 functions as a key regulatory mechanism in their signaling crosstalk.


Sujet(s)
Phagosomes/immunologie , Interactions entre récepteurs/immunologie , Récepteurs du fragment Fc des IgG/immunologie , Récepteur de type Toll-1/immunologie , Récepteur de type Toll-2/immunologie , Animaux , Cytokines/métabolisme , Immunité innée , Immunoglobuline G/immunologie , Membranes intracellulaires/immunologie , Souris , Transport des protéines , Cellules RAW 264.7 , Transduction du signal , Syk kinase/physiologie , Facteur de transcription RelA/métabolisme
8.
Nat Commun ; 12(1): 3519, 2021 06 10.
Article de Anglais | MEDLINE | ID: mdl-34112781

RÉSUMÉ

TLR4 signaling plays key roles in the innate immune response to microbial infection. Innate immune cells encounter different mechanical cues in both health and disease to adapt their behaviors. However, the impact of mechanical sensing signals on TLR4 signal-mediated innate immune response remains unclear. Here we show that TLR4 signalling augments macrophage bactericidal activity through the mechanical sensor Piezo1. Bacterial infection or LPS stimulation triggers assembly of the complex of Piezo1 and TLR4 to remodel F-actin organization and augment phagocytosis, mitochondrion-phagosomal ROS production and bacterial clearance and genetic deficiency of Piezo1 results in abrogation of these responses. Mechanistically, LPS stimulates TLR4 to induce Piezo1-mediated calcium influx and consequently activates CaMKII-Mst1/2-Rac axis for pathogen ingestion and killing. Inhibition of CaMKII or knockout of either Mst1/2 or Rac1 results in reduced macrophage bactericidal activity, phenocopying the Piezo1 deficiency. Thus, we conclude that TLR4 drives the innate immune response via Piezo1 providing critical insight for understanding macrophage mechanophysiology and the host response.


Sujet(s)
Infections bactériennes/immunologie , Immunité innée , Canaux ioniques/métabolisme , Macrophages/immunologie , Phagosomes/métabolisme , Récepteur de type Toll-4/métabolisme , Actines/métabolisme , Animaux , Calcium/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Cytosquelette/génétique , Cytosquelette/métabolisme , Infections à Escherichia coli/immunologie , Transfert d'énergie par résonance de fluorescence , Cellules HEK293 , Facteur de croissance des hépatocytes/génétique , Facteur de croissance des hépatocytes/métabolisme , Humains , Canaux ioniques/génétique , Lipopolysaccharides/pharmacologie , Souris , Souris de lignée C57BL , Neuropeptides/génétique , Neuropeptides/métabolisme , Phagocytose/immunologie , Phagosomes/immunologie , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Espèces réactives de l'oxygène/métabolisme , Serine-threonine kinase-3 , Transduction du signal/génétique , Transduction du signal/immunologie , Récepteur de type Toll-4/immunologie , Protéine G rac1/génétique , Protéine G rac1/métabolisme
9.
Front Immunol ; 12: 662063, 2021.
Article de Anglais | MEDLINE | ID: mdl-33995386

RÉSUMÉ

Phagocytosis is an essential process for the uptake of large (>0.5 µm) particulate matter including microbes and dying cells. Specialized cells in the body perform phagocytosis which is enabled by cell surface receptors that recognize and bind target cells. Professional phagocytes play a prominent role in innate immunity and include macrophages, neutrophils and dendritic cells. These cells display a repertoire of phagocytic receptors that engage the target cells directly, or indirectly via opsonins, to mediate binding and internalization of the target into a phagosome. Phagosome maturation then proceeds to cause destruction and recycling of the phagosome contents. Key subsequent events include antigen presentation and cytokine production to alert and recruit cells involved in the adaptive immune response. Bridging the innate and adaptive immunity, macrophages secrete a broad selection of inflammatory mediators to orchestrate the type and magnitude of an inflammatory response. This review will focus on cytokines produced by NF-κB signaling which is activated by extracellular ligands and serves a master regulator of the inflammatory response to microbes. Macrophages secrete pro-inflammatory cytokines including TNFα, IL1ß, IL6, IL8 and IL12 which together increases vascular permeability and promotes recruitment of other immune cells. The major anti-inflammatory cytokines produced by macrophages include IL10 and TGFß which act to suppress inflammatory gene expression in macrophages and other immune cells. Typically, macrophage cytokines are synthesized, trafficked intracellularly and released in response to activation of pattern recognition receptors (PRRs) or inflammasomes. Direct evidence linking the event of phagocytosis to cytokine production in macrophages is lacking. This review will focus on cytokine output after engagement of macrophage phagocytic receptors by particulate microbial targets. Microbial receptors include the PRRs: Toll-like receptors (TLRs), scavenger receptors (SRs), C-type lectin and the opsonic receptors. Our current understanding of how macrophage receptor stimulation impacts cytokine production is largely based on work utilizing soluble ligands that are destined for endocytosis. We will instead focus this review on research examining receptor ligation during uptake of particulate microbes and how this complex internalization process may influence inflammatory cytokine production in macrophages.


Sujet(s)
Cytokines/immunologie , Macrophages/immunologie , Phagocytes/immunologie , Phagocytes/microbiologie , Transduction du signal/immunologie , Animaux , Antigènes bactériens/immunologie , Cytokines/biosynthèse , Humains , Immunité innée , Souris , Sous-unité p50 de NF-kappa B/immunologie , Phagocytose/immunologie , Phagosomes/immunologie , Phagosomes/microbiologie , Récepteurs de type Toll/immunologie
10.
Front Immunol ; 12: 631714, 2021.
Article de Anglais | MEDLINE | ID: mdl-33959122

RÉSUMÉ

The rapid and efficient phagocytic clearance of apoptotic cells, termed efferocytosis, is a critical mechanism in the maintenance of tissue homeostasis. Removal of apoptotic cells through efferocytosis prevents secondary necrosis and the resultant inflammation caused by the release of intracellular contents. The importance of efferocytosis in homeostasis is underscored by the large number of inflammatory and autoimmune disorders, including atherosclerosis and systemic lupus erythematosus, that are characterized by defective apoptotic cell clearance. Although mechanistically similar to the phagocytic clearance of pathogens, efferocytosis differs from phagocytosis in that it is immunologically silent and induces a tissue repair response. Efferocytes face unique challenges resulting from the internalization of apoptotic cells, including degradation of the apoptotic cell, dealing with the extra metabolic load imposed by the processing of apoptotic cell contents, and the coordination of an anti-inflammatory, pro-tissue repair response. This review will discuss recent advances in our understanding of the cellular response to apoptotic cell uptake, including trafficking of apoptotic cell cargo and antigen presentation, signaling and transcriptional events initiated by efferocytosis, the coordination of an anti-inflammatory response and tissue repair, unique cellular metabolic responses and the role of efferocytosis in host defense. A better understanding of how efferocytic cells respond to apoptotic cell uptake will be critical in unraveling the complex connections between apoptotic cell removal and inflammation resolution and maintenance of tissue homeostasis.


Sujet(s)
Apoptose , Phagocytose , Présentation d'antigène , Apoptose/immunologie , Régulation de l'expression des gènes , Homéostasie , Humains , Inflammation/immunologie , Phagocytes/immunologie , Phagocytes/métabolisme , Phagocytose/immunologie , Phagosomes/immunologie , Phagosomes/métabolisme , Transduction du signal
11.
mSphere ; 6(3)2021 05 05.
Article de Anglais | MEDLINE | ID: mdl-33952660

RÉSUMÉ

Mycobacterium tuberculosis infections claim more than a million lives each year, and better treatments or vaccines are required. A crucial pathogenicity factor is translocation from phagolysosomes to the cytosol upon phagocytosis by macrophages. Translocation from the phagolysosome to the cytosol is an ESX-1-dependent process, as previously shown in vitro Here, we show that in vivo, mycobacteria also translocate to the cytosol but mainly when host immunity is compromised. We observed only low numbers of cytosolic bacilli in mice, armadillos, zebrafish, and patient material infected with M. tuberculosis, M. marinum, or M. leprae In contrast, when innate or adaptive immunity was compromised, as in severe combined immunodeficiency (SCID) or interleukin-1 receptor 1 (IL-1R1)-deficient mice, significant numbers of cytosolic M. tuberculosis bacilli were detected in the lungs of infected mice. Taken together, in vivo, translocation to the cytosol of M. tuberculosis is controlled by adaptive immune responses as well as IL-1R1-mediated signals.IMPORTANCE For decades, Mycobacterium tuberculosis has been one of the deadliest pathogens known. Despite infecting approximately one-third of the human population, no effective treatment or vaccine is available. A crucial pathogenicity factor is subcellular localization, as M. tuberculosis can translocate from phagolysosome to the cytosol in macrophages. The situation in vivo is more complicated. In this study, we establish that high-level cytosolic escape of mycobacteria can indeed occur in vivo but mainly when host resistance is compromised. The IL-1 pathway is crucial for the control of the number of cytosolic mycobacteria. The establishment that immune signals result in the clearance of cells containing cytosolic mycobacteria connects two important fields, cell biology and immunology, which is vital for the understanding of the pathology of M. tuberculosis.


Sujet(s)
Cytosol/microbiologie , Mycobacterium/immunologie , Mycobacterium/pathogénicité , Phagosomes/microbiologie , Récepteurs à l'interleukine-1/génétique , Récepteurs à l'interleukine-1/immunologie , Transduction du signal/immunologie , Animaux , Tatous/microbiologie , Translocation bactérienne , Cytosol/immunologie , Femelle , Humains , Lèpre/microbiologie , Mâle , Souris , Souris de lignée BALB C , Souris SCID , Mycobacterium/classification , Phagosomes/immunologie , Peau/microbiologie , Peau/anatomopathologie , Cellules THP-1 , Danio zébré
12.
Front Immunol ; 12: 659533, 2021.
Article de Anglais | MEDLINE | ID: mdl-33868308

RÉSUMÉ

Phagocytosis is a receptor-mediated process used by cells to engulf a wide variety of particulates, including microorganisms and apoptotic cells. Many of the proteins involved in this highly orchestrated process are post-translationally modified with lipids as a means of regulating signal transduction, membrane remodeling, phagosome maturation and other immunomodulatory functions of phagocytes. S-acylation, generally referred to as S-palmitoylation, is the post-translational attachment of fatty acids to a cysteine residue exposed topologically to the cytosol. This modification is reversible due to the intrinsically labile thioester bond between the lipid and sulfur atom of cysteine, and thus lends itself to a variety of regulatory scenarios. Here we present an overview of a growing number of S-acylated proteins known to regulate phagocytosis and phagosome biology in macrophages.


Sujet(s)
Macrophages/immunologie , Phagocytose/immunologie , Phagosomes/immunologie , Maturation post-traductionnelle des protéines , Protéome/immunologie , Acylation , Animaux , Humains , Macrophages/métabolisme , Phagosomes/métabolisme , Protéome/métabolisme , Protéomique/méthodes , Transduction du signal/immunologie
13.
Am J Respir Cell Mol Biol ; 65(2): 176-188, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33848212

RÉSUMÉ

Macrophages undergo profound metabolic reprogramming to join key immunoregulatory functions, which can be initiated by pattern recognition receptors. TREM2 (triggering receptor expressed on myeloid cells 2), a macrophage phagocytic receptor, plays pivotal roles in sepsis by enhancing bacterial clearance, which is associated with regulation of reactive oxygen species (ROS) production. However, how intracellular ROS participate in TREM2-mediated bactericidal activity remains unclear. This study was designed to investigate the organelle source and biological activity of ROS in the context of TREM2-mediated immune defense during Escherichiacoli infection. Bone marrow-derived macrophages (BMDMs) were transfected with TREM2-overexpressing adenoviruses or control viruses and challenged with E. coli. The BMDMs were administered to mouse models with local E. coli infection. In addition, monocytic TREM2 expression, NOX2 concentrations, and pyroptosis were detected in patients with bacterial sepsis. General ROS production was found to be comparable between TREM2-overexpressing and control BMDMs upon E. coli challenge. The deficiency of Nox2 led to impaired phagosome degradation and lack of bactericidal ability and abolished TREM2-mediated protective activity against pulmonary E. coli infection. Overexpression of TREM2 suppressed mitochondrial ROS generation, inhibited NLRP3/caspase-1 inflammasome activation, and finally protected BMDMs from gasdermin D-mediated pyroptosis during pulmonary E. coli infection. The protective role of TREM2 was further confirmed in mice with abdominal E. coli infection. Moreover, monocytic TREM2 expression was positively correlated with NOX2 concentrations and negatively correlated with pyroptosis and disease severity in patients with bacterial sepsis. Collectively, TREM2 controls macrophage immune functions by fine-tuning ROS generation and enhances the host defense against bacterial infection. Our data suggest that TREM2 is a promising candidate target for sepsis therapy.


Sujet(s)
Cellules de la moelle osseuse/immunologie , Infections à Escherichia coli/immunologie , Escherichia coli/immunologie , Macrophages/immunologie , Glycoprotéines membranaires/immunologie , Pneumopathie bactérienne/immunologie , Récepteurs immunologiques/immunologie , Animaux , Cellules de la moelle osseuse/anatomopathologie , Infections à Escherichia coli/génétique , Régulation de l'expression des gènes/immunologie , Macrophages/anatomopathologie , Glycoprotéines membranaires/génétique , Souris , Souris knockout , NADPH Oxidase 2/génétique , NADPH Oxidase 2/immunologie , Phagosomes/génétique , Phagosomes/immunologie , Pneumopathie bactérienne/génétique , Pneumopathie bactérienne/anatomopathologie , Récepteurs immunologiques/génétique
14.
Infect Immun ; 89(7): e0000921, 2021 06 16.
Article de Anglais | MEDLINE | ID: mdl-33875473

RÉSUMÉ

Leishmaniasis, a debilitating disease with clinical manifestations ranging from self-healing ulcers to life-threatening visceral pathologies, is caused by protozoan parasites of the Leishmania genus. These professional vacuolar pathogens are transmitted by infected sand flies to mammalian hosts as metacyclic promastigotes and are rapidly internalized by various phagocyte populations. Classical monocytes are among the first myeloid cells to migrate to infection sites. Recent evidence shows that recruitment of these cells contributes to parasite burden and the establishment of chronic disease. However, the nature of Leishmania-inflammatory monocyte interactions during the early stages of host infection has not been well investigated. Here, we aimed to assess the impact of Leishmania donovani metacyclic promastigotes on antimicrobial responses within these cells. Our data showed that inflammatory monocytes are readily colonized by L. donovani metacyclic promastigotes, while infection with Escherichia coli is efficiently cleared. Upon internalization, metacyclic promastigotes inhibited superoxide production at the parasitophorous vacuole (PV) through a mechanism involving exclusion of NADPH oxidase subunits gp91phox and p47phox from the PV membrane. Moreover, we observed that unlike phagosomes enclosing zymosan particles, vacuoles containing parasites acidify poorly. Interestingly, whereas the parasite surface coat virulence glycolipid lipophosphoglycan (LPG) was responsible for the inhibition of PV acidification, impairment of the NADPH oxidase assembly was independent of LPG and GP63. Collectively, these observations indicate that permissiveness of inflammatory monocytes to L. donovani may thus be related to the ability of this parasite to impair the microbicidal properties of phagosomes.


Sujet(s)
Interactions hôte-parasite , Leishmania donovani/immunologie , Leishmaniose viscérale/immunologie , Leishmaniose viscérale/parasitologie , Monocytes/immunologie , Monocytes/parasitologie , Phagosomes/immunologie , Phagosomes/parasitologie , Glycosphingolipides/métabolisme , Interactions hôte-parasite/immunologie , Leishmania donovani/métabolisme , Leishmania donovani/pathogénicité , Monocytes/métabolisme , NADPH oxidase/métabolisme , Virulence , Facteurs de virulence
15.
Front Immunol ; 12: 662987, 2021.
Article de Anglais | MEDLINE | ID: mdl-33815423

RÉSUMÉ

Hijacking the autophagic machinery is a key mechanism through which invasive pathogens such as Staphylococcus aureus replicate in their host cells. We have previously demonstrated that the bacteria replicate in phagosomes labeled with the autophagic protein LC3, before escaping to the cytoplasm. Here, we show that the Ca2+-dependent PKCα binds to S. aureus-containing phagosomes and that α-hemolysin, secreted by S. aureus, promotes this recruitment of PKCα to phagosomal membranes. Interestingly, the presence of PKCα prevents the association of the autophagic protein LC3. Live cell imaging experiments using the PKC activity reporter CKAR reveal that treatment of cells with S. aureus culture supernatants containing staphylococcal secreted factors transiently activates PKC. Functional studies reveal that overexpression of PKCα causes a marked inhibition of bacterial replication. Taken together, our data identify enhancing PKCα activity as a potential approach to inhibit S. aureus replication in mammalian cells.


Sujet(s)
Autophagie , Interactions hôte-pathogène , Phagosomes/métabolisme , Protein kinase C-alpha/métabolisme , Infections à staphylocoques/étiologie , Infections à staphylocoques/métabolisme , Staphylococcus aureus/physiologie , Animaux , Autophagie/immunologie , Cellules CHO , Lignée cellulaire , Cellules cultivées , Cricetulus , Prédisposition aux maladies , Technique d'immunofluorescence , Gènes rapporteurs , Interactions hôte-pathogène/immunologie , Modèles biologiques , Phagosomes/immunologie , Protein kinase C-alpha/génétique
16.
Front Immunol ; 12: 636078, 2021.
Article de Anglais | MEDLINE | ID: mdl-33717183

RÉSUMÉ

Following phagocytosis, the nascent phagosome undergoes maturation to become a phagolysosome with an acidic, hydrolytic, and often oxidative lumen that can efficiently kill and digest engulfed microbes, cells, and debris. The fusion of phagosomes with lysosomes is a principal driver of phagosomal maturation and is targeted by several adapted intracellular pathogens. Impairment of this process has significant consequences for microbial infection, tissue inflammation, the onset of adaptive immunity, and disease. Given the importance of phagosome-lysosome fusion to phagocyte function and the many virulence factors that target it, it is unsurprising that multiple molecular pathways have evolved to mediate this essential process. While the full range of these pathways has yet to be fully characterized, several pathways involving proteins such as members of the Rab GTPases, tethering factors and SNAREs have been identified. Here, we summarize the current state of knowledge to clarify the ambiguities in the field and construct a more comprehensive phagolysosome formation model. Lastly, we discuss how other cellular pathways help support phagolysosome biogenesis and, consequently, phagocyte function.


Sujet(s)
Lysosomes/métabolisme , Fusion membranaire , Phagocytes/métabolisme , Phagocytose , Phagosomes/métabolisme , Animaux , Autophagie , Humains , Lysosomes/immunologie , Phagocytes/immunologie , Phagosomes/immunologie , Protéines SNARE/métabolisme , Protéines G rab/métabolisme
17.
mSphere ; 6(1)2021 01 06.
Article de Anglais | MEDLINE | ID: mdl-33408238

RÉSUMÉ

Bryan D. Bryson works in the field of biological engineering with a specific interest in host-mycobacterium interactions. In this mSphere of Influence article, he reflects on how "IRG1 and inducible nitric oxide synthase act redundantly with other interferon-gamma-induced factors to restrict intracellular replication of Legionella pneumophila" by Price and colleagues (J. V. Price, D. Russo, D. X. Ji, R. A. Chavez, et al., mBio 10:e02629-19, 2019, https://doi.org/10.1128/mBio.02629-19) made an impact on him by reinforcing the complexity of intracellular pathogen control.


Sujet(s)
Interactions hôte-pathogène , Immunité innée/génétique , Interféron gamma/immunologie , Legionella pneumophila/immunologie , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/immunologie , Humains , Legionella pneumophila/pathogénicité , Narration , Nitric oxide synthase type II , Phagosomes/immunologie , Phagosomes/microbiologie , Transport des protéines
18.
FEBS J ; 288(5): 1412-1433, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-32757358

RÉSUMÉ

Phagocytosis is an essential mechanism for immunity and homeostasis, performed by a subset of cells known as phagocytes. Upon target engulfment, de novo formation of specialized compartments termed phagosomes takes place. Phagosomes then undergo a series of fusion and fission events as they interact with the endolysosomal system and other organelles, in a dynamic process known as phagosome maturation. Because phagocytes play a key role in tissue patrolling and immune surveillance, phagosome maturation is associated with signaling pathways that link phagocytosis to antigen presentation and the development of adaptive immune responses. In addition, and depending on the nature of the cargo, phagosome integrity may be compromised, triggering additional cellular mechanisms including inflammation and autophagy. Upon completion of maturation, phagosomes enter a recently described phase: phagosome resolution, where catabolites from degraded cargo are metabolized, phagosomes are resorbed, and vesicles of phagosomal origin are recycled. Finally, phagocytes return to homeostasis and become ready for a new round of phagocytosis. Altogether, phagosome maturation and resolution encompass a series of dynamic events and organelle crosstalk that can be measured by biochemical, imaging, photoluminescence, cytometric, and immune-based assays that will be described in this guide.


Sujet(s)
Endosomes/immunologie , Lysosomes/immunologie , Phagocytes/immunologie , Phagocytose , Phagosomes/immunologie , Immunité acquise , Animaux , Présentation d'antigène , Autophagie/génétique , Autophagie/immunologie , Endosomes/métabolisme , Endosomes/ultrastructure , Humains , Immunité innée , Dosage immunologique , Surveillance immunologique , Inflammation , Lysosomes/métabolisme , Lysosomes/ultrastructure , Techniques de sonde moléculaire , Phagocytes/métabolisme , Phagocytes/ultrastructure , Phagosomes/métabolisme , Phagosomes/ultrastructure , Transduction du signal
19.
Nat Immunol ; 22(2): 140-153, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33349708

RÉSUMÉ

Type 1 conventional dendritic (cDC1) cells are necessary for cross-presentation of many viral and tumor antigens to CD8+ T cells. cDC1 cells can be identified in mice and humans by high expression of DNGR-1 (also known as CLEC9A), a receptor that binds dead-cell debris and facilitates XP of corpse-associated antigens. Here, we show that DNGR-1 is a dedicated XP receptor that signals upon ligand engagement to promote phagosomal rupture. This allows escape of phagosomal contents into the cytosol, where they access the endogenous major histocompatibility complex class I antigen processing pathway. The activity of DNGR-1 maps to its signaling domain, which activates SYK and NADPH oxidase to cause phagosomal damage even when spliced into a heterologous receptor and expressed in heterologous cells. Our data reveal the existence of innate immune receptors that couple ligand binding to endocytic vesicle damage to permit MHC class I antigen presentation of exogenous antigens and to regulate adaptive immunity.


Sujet(s)
Présentation d'antigène , Cross-priming , Cellules dendritiques/métabolisme , Lectines de type C/métabolisme , Phagosomes/métabolisme , Récepteurs immunologiques/métabolisme , Récepteur mitogène/métabolisme , Lymphocytes T/métabolisme , Animaux , Mort cellulaire , Techniques de coculture , Cellules dendritiques/immunologie , Cellules HEK293 , Antigènes d'histocompatibilité de classe I/métabolisme , Humains , Lectines de type C/génétique , Ligands , Souris , NADPH oxidase/métabolisme , Phagosomes/génétique , Phagosomes/immunologie , Phosphorylation , Cellules RAW 264.7 , Espèces réactives de l'oxygène/métabolisme , Récepteurs immunologiques/génétique , Récepteur mitogène/génétique , Transduction du signal , Syk kinase/métabolisme , Lymphocytes T/immunologie
20.
PLoS Pathog ; 16(9): e1008328, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32936835

RÉSUMÉ

Candida albicans cells depend on the energy derived from amino acid catabolism to induce and sustain hyphal growth inside phagosomes of engulfing macrophages. The concomitant deamination of amino acids is thought to neutralize the acidic microenvironment of phagosomes, a presumed requisite for survival and initiation of hyphal growth. Here, in contrast to an existing model, we show that mitochondrial-localized NAD+-dependent glutamate dehydrogenase (GDH2) catalyzing the deamination of glutamate to α-ketoglutarate, and not the cytosolic urea amidolyase (DUR1,2), accounts for the observed alkalization of media when amino acids are the sole sources of carbon and nitrogen. C. albicans strains lacking GDH2 (gdh2-/-) are viable and do not extrude ammonia on amino acid-based media. Environmental alkalization does not occur under conditions of high glucose (2%), a finding attributable to glucose-repression of GDH2 expression and mitochondrial function. Consistently, inhibition of oxidative phosphorylation or mitochondrial translation by antimycin A or chloramphenicol, respectively, prevents alkalization. GDH2 expression and mitochondrial function are derepressed as glucose levels are lowered from 2% (~110 mM) to 0.2% (~11 mM), or when glycerol is used as primary carbon source. Using time-lapse microscopy, we document that gdh2-/- cells survive, filament and escape from primary murine macrophages at rates indistinguishable from wildtype. In intact hosts, such as in fly and murine models of systemic candidiasis, gdh2-/- mutants are as virulent as wildtype. Thus, although Gdh2 has a critical role in central nitrogen metabolism, Gdh2-catalyzed deamination of glutamate is surprisingly dispensable for escape from macrophages and virulence. Consistently, using the pH-sensitive dye (pHrodo), we observed no significant difference between wildtype and gdh2-/- mutants in phagosomal pH modulation. Following engulfment of fungal cells, the phagosomal compartment is rapidly acidified and hyphal growth initiates and sustained under consistently acidic conditions within phagosomes. Together, our results demonstrate that amino acid-dependent alkalization is not essential for hyphal growth, survival in macrophages and hosts. An accurate understanding of the microenvironment within macrophage phagosomes and the metabolic events underlying the survival of phagocytized C. albicans cells and their escape are critical to understanding the host-pathogen interactions that ultimately determine the pathogenic outcome.


Sujet(s)
Candida albicans/immunologie , Candidose/immunologie , Drosophila melanogaster/immunologie , Glutamate dehydrogenase/métabolisme , Macrophages/immunologie , Acides aminés/génétique , Acides aminés/métabolisme , Animaux , Candida albicans/pathogénicité , Candidose/métabolisme , Candidose/microbiologie , Drosophila melanogaster/métabolisme , Drosophila melanogaster/microbiologie , Femelle , Protéines fongiques/génétique , Protéines fongiques/métabolisme , Glutamate dehydrogenase/génétique , Interactions hôte-pathogène , Concentration en ions d'hydrogène , Macrophages/microbiologie , Souris , Souris de lignée C57BL , Azote , Phagosomes/immunologie , Phagosomes/métabolisme , Phagosomes/microbiologie , Virulence
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE