Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 292
Filtrer
1.
Sci Adv ; 10(36): eadl6162, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39241063

RÉSUMÉ

Carbapenem-resistant Klebsiella pneumoniae (CRKP) causes Gram-negative lung infections and fatal pneumonic sepsis for which limited therapeutic options are available. The lungs are densely innervated by nociceptor sensory neurons that mediate breathing, cough, and bronchoconstriction. The role of nociceptors in defense against Gram-negative lung pathogens is unknown. Here, we found that lung-innervating nociceptors promote CRKP pneumonia and pneumonic sepsis. Ablation of nociceptors in mice increased lung CRKP clearance, suppressed trans-alveolar dissemination of CRKP, and protected mice from hypothermia and death. Furthermore, ablation of nociceptors enhanced the recruitment of neutrophils and Ly6Chi monocytes and cytokine induction. Depletion of Ly6Chi monocytes, but not of neutrophils, abrogated lung and extrapulmonary CRKP clearance in ablated mice, suggesting that Ly6Chi monocytes are a critical cellular population to regulate pneumonic sepsis. Further, neuropeptide calcitonin gene-related peptide suppressed the induction of reactive oxygen species in Ly6Chi monocytes and their CRKP-killing abilities. Targeting nociceptor signaling could be a therapeutic approach for treating multidrug-resistant Gram-negative infection and pneumonic sepsis.


Sujet(s)
Peptide relié au gène de la calcitonine , Carbapénèmes , Infections à Klebsiella , Klebsiella pneumoniae , Poumon , Nocicepteurs , Sepsie , Animaux , Klebsiella pneumoniae/physiologie , Souris , Infections à Klebsiella/microbiologie , Sepsie/métabolisme , Sepsie/microbiologie , Poumon/microbiologie , Poumon/métabolisme , Carbapénèmes/pharmacologie , Peptide relié au gène de la calcitonine/métabolisme , Nocicepteurs/métabolisme , Monocytes/métabolisme , Cellules réceptrices sensorielles/métabolisme , Granulocytes neutrophiles/métabolisme , Modèles animaux de maladie humaine , Antigènes Ly/métabolisme , Espèces réactives de l'oxygène/métabolisme , Pneumopathie bactérienne/microbiologie , Pneumopathie bactérienne/métabolisme , Pneumopathie bactérienne/anatomopathologie , Souris de lignée C57BL
2.
J Clin Invest ; 2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39207863

RÉSUMÉ

Intestinal dysmotility syndromes have been epidemiologically associated with several antecedent bacterial and viral infections. To model this phenotype, we previously infected mice with the neurotropic flavivirus, West Nile Virus (WNV) and demonstrated intestinal transit defects. Here, we find that within one week of WNV infection, enteric neurons and glia become damaged, resulting in sustained reductions of neuronal cells and their networks of connecting fibers. Using cell-depleting antibodies, adoptive transfer experiments, and mice lacking specific immune cells or immune functions, we show that infiltrating WNV-specific CD4+ and CD8+ T cells damage the enteric nervous system (ENS) and glia, which leads to intestinal dysmotility; these T cells use multiple and redundant effector functions including perforin and Fas ligand. In comparison, WNV-triggered ENS injury and intestinal dysmotility appears to not require infiltrating monocytes and damage may be limited by resident muscularis macrophages. Overall, our experiments support a model whereby antigen specific T cell subsets and their effector molecules responding to WNV infection direct immune pathology against enteric neurons and supporting glia that results in intestinal dysmotility.

3.
J Virol ; 98(9): e0117924, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39207134

RÉSUMÉ

Cardiovascular manifestations of coronavirus disease 2019 (COVID-19) include myocardial injury, heart failure, and myocarditis and are associated with long-term disability and mortality. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA and antigens are found in the myocardium of COVID-19 patients, and human cardiomyocytes are susceptible to infection in cell or organoid cultures. While these observations raise the possibility that cardiomyocyte infection may contribute to the cardiac sequelae of COVID-19, a causal relationship between cardiomyocyte infection and myocardial dysfunction and pathology has not been established. Here, we generated a mouse model of cardiomyocyte-restricted infection by selectively expressing human angiotensin-converting enzyme 2 (hACE2), the SARS-CoV-2 receptor, in cardiomyocytes. Inoculation of Myh6-Cre Rosa26loxP-STOP-loxP-hACE2 mice with an ancestral, non-mouse-adapted strain of SARS-CoV-2 resulted in viral replication within the heart, accumulation of macrophages, and moderate left ventricular (LV) systolic dysfunction. Cardiac pathology in this model was transient and resolved with viral clearance. Blockade of monocyte trafficking reduced macrophage accumulation, suppressed the development of LV systolic dysfunction, and promoted viral clearance in the heart. These findings establish a mouse model of SARS-CoV-2 cardiomyocyte infection that recapitulates features of cardiac dysfunctions of COVID-19 and suggests that both viral replication and resultant innate immune responses contribute to cardiac pathology.IMPORTANCEHeart involvement after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection occurs in multiple ways and is associated with worse outcomes in coronavirus disease 2019 (COVID-19) patients. It remains unclear if cardiac disease is driven by primary infection of the heart or immune response to the virus. SARS-CoV-2 is capable of entering contractile cells of the heart in a culture dish. However, it remains unclear how such infection affects the function of the heart in the body. Here, we designed a mouse in which only heart muscle cells can be infected with a SARS-CoV-2 strain to study cardiac infection in isolation from other organ systems. In our model, infected mice show viral infection, worse function, and accumulation of immune cells in the heart. A subset of immune cells facilitates such worsening heart function. As this model shows features similar to those observed in patients, it may be useful for understanding the heart disease that occurs as a part of COVID-19.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Modèles animaux de maladie humaine , Monocytes , Myocytes cardiaques , SARS-CoV-2 , Animaux , COVID-19/immunologie , COVID-19/virologie , COVID-19/anatomopathologie , Souris , Myocytes cardiaques/virologie , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Monocytes/immunologie , Monocytes/virologie , Humains , Macrophages/virologie , Macrophages/immunologie , Réplication virale , Myocarde/anatomopathologie , Myocarde/immunologie , Dysfonction ventriculaire gauche/virologie , Dysfonction ventriculaire gauche/physiopathologie , Dysfonction ventriculaire gauche/anatomopathologie
4.
J Mol Biol ; 436(20): 168734, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39097184

RÉSUMÉ

The antibiotic roseoflavin is a riboflavin (vitamin B2) analog. One step of the roseoflavin biosynthetic pathway is catalyzed by the phosphatase RosC, which dephosphorylates 8-demethyl-8-amino-riboflavin-5'-phosphate (AFP) to 8-demethyl-8-amino-riboflavin (AF). RosC also catalyzes the potentially cell-damaging dephosphorylation of the AFP analog riboflavin-5'-phosphate also called "flavin mononucleotide" (FMN), however, with a lower efficiency. We performed X-ray structural analyses and mutagenesis studies on RosC from Streptomyces davaonensis to understand binding of the flavin substrates, the distinction between AFP and FMN and the catalytic mechanism of this enzyme. This work is the first structural analysis of an AFP phosphatase. Each monomer of the RosC dimer consists of an α/ß-fold core, which is extended by three specific elongated strand-to-helix sections and a specific N-terminal helix. Altogether these segments envelope the flavin thereby forming a novel flavin-binding site. We propose that distinction between AFP and FMN is provided by substrate-induced rigidification of the four RosC specific supplementary segments mentioned above and by an interaction between the amino group at C8 of AFP and the ß-carboxylate of D166. This key amino acid is involved in binding the ring system of AFP and positioning its ribitol phosphate part. Accordingly, site-specific exchanges at D166 disturbed the active site geometry of the enzyme and drastically reduced the catalytic activity. Based on the structure of the catalytic core we constructed a whole series of RosC variants but a disturbing, FMN dephosphorylating "killer enzyme", was not generated.


Sujet(s)
Flavine mononucléotide , Riboflavine , Streptomyces , Streptomyces/génétique , Streptomyces/métabolisme , Streptomyces/enzymologie , Riboflavine/analogues et dérivés , Riboflavine/biosynthèse , Riboflavine/métabolisme , Flavine mononucléotide/métabolisme , Cristallographie aux rayons X , Phosphoric monoester hydrolases/métabolisme , Phosphoric monoester hydrolases/génétique , Phosphorylation , Modèles moléculaires , Sites de fixation , Conformation des protéines , Spécificité du substrat
5.
Mucosal Immunol ; 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39127259

RÉSUMÉ

Respiratory syncytial virus (RSV) can cause severe lower respiratory tract infections. Understanding why some individuals get more serious disease may help with diagnosis and treatment. One possible risk factor underlying severe disease is bacterial exposure before RSV infection. Bacterial exposure has been associated with increased respiratory viral-induced disease severity but the mechanism remains unknown. Respiratory bacterial infections or exposure to their pathogen associated molecular patterns (PAMPs) trigger innate immune inflammation, characterised by neutrophil and inflammatory monocyte recruitment and the production of inflammatory cytokines. We hypothesise that these changes to the lung environment alter the immune response and disease severity during subsequent RSV infection. To test this, we intranasally exposed mice to LPS, LTA or Acinetobacter baumannii (an airway bacterial pathogen) before RSV infection and observed an early induction of disease, measured by weight loss, at days 1-3 after infection. Neutrophils or inflammatory monocytes were not responsible for driving this exacerbated weight loss. Instead, exacerbated disease was associated with increased IL-1α and TNF-α, which orchestrated the recruitment of innate immune cells into the lung. This study shows that exposure to bacterial PAMPs prior to RSV infection increases the expression of IL-1α and TNF-α, which dysregulate the immune response resulting in exacerbated disease.

6.
Am J Pathol ; 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39117108

RÉSUMÉ

Monocyte patrolling of the vasculature has been ascribed primarily to the non-classical monocyte subset. However, a recent study of the glomerular microvasculature provided evidence that both classical and non-classical monocytes undergo periods of intravascular retention and migration. Despite this, whether these subsets contribute differentially to acute glomerular inflammation is unknown. This study used glomerular multiphoton intravital microscopy to investigate the capacity of classical and non-classical monocytes to patrol the glomerular microvasculature and promote acute, neutrophil-dependent glomerular inflammation. In imaging experiments in monocyte reporter Cx3cr1gfp/+ mice, co-staining with anti-Ly6B or anti-Ly6C revealed that both non-classical monocytes [CX3C chemokine receptor 1-green fluorescent protein positive (CX3CR1-GFP+)] and classical monocytes (CX3CR1-GFP+ and Ly6B+ or Ly6C+) underwent prolonged (>10 minutes) retention and migration in the glomerular microvasculature. On induction of acute glomerulonephritis, in these behaviors were increased in classical but not non-classical monocytes. Using non-classical monocyte-deficient Csf1rCreNr4a1fl/fl mice, or anti-CCR2 to deplete classical monocytes, the removal of either subset reduced neutrophil retention and activation in acutely inflamed glomeruli, while the depletion of both subsets, via anti-CCR2 treatment in Csf1rCreNr4a1fl/fl mice, led to further reductions in neutrophil activity. In contrast, in a model of CD4+ T cell-dependent glomerulonephritis, the depletion of either monocyte subset failed to alter neutrophil responses. These findings indicate that both classical and non-classical monocytes patrol the glomerular microvasculature and promote neutrophil responses in acutely inflamed glomeruli.

7.
bioRxiv ; 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39005354

RÉSUMÉ

Bacterial vaginosis (BV) is a dysbiosis of the vaginal microbiome that is prevalent in reproductive-age women worldwide. Adverse outcomes associated with BV include an increased risk of sexually acquired Human Immunodeficiency Virus (HIV), yet the immunological mechanisms underlying this association are not well understood. To investigate BV driven changes to cervicovaginal tract (CVT) and circulating T cell phenotypes, participants with or without BV provided vaginal tract (VT) and ectocervical (CX) tissue biopsies and peripheral blood mononuclear cells (PBMC). Immunofluorescence analysis of genital mucosal tissues revealed a reduced density of CD3+CD4+CCR5+ cells in the VT lamina propria of individuals with compared to those without BV (median 243.8 cells/mm2 BV- vs 106.9 cells/mm2 BV+, p=0.043). High-parameter flow cytometry of VT biopsies revealed an increased frequency in individuals with compared to those without BV of dysfunctional CD39+ conventional CD4+ T cells (Tconv) (median frequency 15% BV- vs 30% BV+, padj=0.0331) and tissue-resident CD69+CD103+ Tconv (median frequency 24% BV- vs 38% BV+, padj=0.0061), previously reported to be implicated in HIV acquisition and replication. Our data suggests that BV elicits diverse and complex VT T cell alterations and expands on potential immunological mechanisms that may promote adverse outcomes including HIV susceptibility.

8.
ACS Infect Dis ; 10(8): 2939-2949, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-38920250

RÉSUMÉ

The riboflavin analogues, roseoflavin and 8-aminoriboflavin, inhibit malaria parasite proliferation by targeting riboflavin utilization. To determine their mechanism of action, we generated roseoflavin-resistant parasites by in vitro evolution. Relative to wild-type, these parasites were 4-fold resistant to roseoflavin and cross-resistant to 8-aminoriboflavin. Whole genome sequencing of the resistant parasites revealed a missense mutation leading to an amino acid change (L672H) in the gene coding for a putative flavokinase (PfFK), the enzyme responsible for converting riboflavin into the cofactor flavin mononucleotide (FMN). To confirm that the L672H mutation is responsible for the phenotype, we generated parasites with the missense mutation incorporated into the PfFK gene. The IC50 values for roseoflavin and 8-aminoriboflavin against the roseoflavin-resistant parasites created through in vitro evolution were indistinguishable from those against parasites in which the missense mutation was introduced into the native PfFK. We also generated two parasite lines episomally expressing GFP-tagged versions of either the wild-type or mutant forms of PfFK. We found that PfFK-GFP localizes to the parasite cytosol and that immunopurified PfFK-GFP phosphorylated riboflavin, roseoflavin, and 8-aminoriboflavin. The L672H mutation increased the KM for roseoflavin, explaining the resistance phenotype. Mutant PfFK is no longer capable of phosphorylating 8-aminoriboflavin, but its antiplasmodial activity against resistant parasites can still be antagonized by increasing the extracellular concentration of riboflavin, consistent with it also inhibiting parasite growth through competitive inhibition of PfFK. Our findings, therefore, are consistent with roseoflavin and 8-aminoriboflavin inhibiting parasite proliferation by inhibiting riboflavin phosphorylation and via the generation of toxic flavin cofactor analogues.


Sujet(s)
Antipaludiques , Résistance aux substances , Phosphotransferases (Alcohol Group Acceptor) , Plasmodium falciparum , Riboflavine , Plasmodium falciparum/effets des médicaments et des substances chimiques , Plasmodium falciparum/génétique , Plasmodium falciparum/enzymologie , Riboflavine/pharmacologie , Riboflavine/analogues et dérivés , Antipaludiques/pharmacologie , Résistance aux substances/génétique , Phosphotransferases (Alcohol Group Acceptor)/génétique , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Mutation faux-sens , Humains , Paludisme à Plasmodium falciparum/parasitologie , Mutation
9.
Allergy ; 79(8): 2157-2172, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38924546

RÉSUMÉ

BACKGROUND: Respiratory syncytial virus (RSV) infection in infants is a major cause of viral bronchiolitis and hospitalisation. We have previously shown in a murine model that ongoing infection with the gut helminth Heligmosomoides polygyrus protects against RSV infection through type I interferon (IFN-I) dependent reduction of viral load. Yet, the cellular basis for this protection has remained elusive. Given that recruitment of mononuclear phagocytes to the lung is critical for early RSV infection control, we assessed their role in this coinfection model. METHODS: Mice were infected by oral gavage with H. polygyrus. Myeloid immune cell populations were assessed by flow cytometry in lung, blood and bone marrow throughout infection and after secondary infection with RSV. Monocyte numbers were depleted by anti-CCR2 antibody or increased by intravenous transfer of enriched monocytes. RESULTS: H. polygyrus infection induces bone marrow monopoiesis, increasing circulatory monocytes and lung mononuclear phagocytes in a IFN-I signalling dependent manner. This expansion causes enhanced lung mononuclear phagocyte counts early in RSV infection that may contribute to the reduction of RSV load. Depletion or supplementation of circulatory monocytes prior to RSV infection confirms that these are both necessary and sufficient for helminth induced antiviral protection. CONCLUSIONS: H. polygyrus infection induces systemic monocytosis contributing to elevated mononuclear phagocyte numbers in the lung. These cells are central to an anti-viral effect that reduces the peak viral load in RSV infection. Treatments to promote or modulate these cells may provide novel paths to control RSV infection in high risk individuals.


Sujet(s)
Modèles animaux de maladie humaine , Monocytes , Infections à virus respiratoire syncytial , Charge virale , Animaux , Infections à virus respiratoire syncytial/immunologie , Souris , Monocytes/immunologie , Nematospiroides dubius/immunologie , Poumon/immunologie , Poumon/virologie , Infections à Strongylida/immunologie , Virus respiratoires syncytiaux/immunologie , Interféron de type I/métabolisme
10.
Nat Cancer ; 5(8): 1227-1249, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38741011

RÉSUMÉ

Cancer immunotherapy with chimeric antigen receptor (CAR) T cells can cause immune effector cell-associated neurotoxicity syndrome (ICANS). However, the molecular mechanisms leading to ICANS are not well understood. Here we examined the role of microglia using mouse models and cohorts of individuals with ICANS. CD19-directed CAR (CAR19) T cell transfer in B cell lymphoma-bearing mice caused microglia activation and neurocognitive deficits. The TGFß-activated kinase-1 (TAK1)-NF-κB-p38 MAPK pathway was activated in microglia after CAR19 T cell transfer. Pharmacological TAK1 inhibition or genetic Tak1 deletion in microglia using Cx3cr1CreER:Tak1fl/fl mice resulted in reduced microglia activation and improved neurocognitive activity. TAK1 inhibition allowed for potent CAR19-induced antilymphoma effects. Individuals with ICANS exhibited microglia activation in vivo when studied by translocator protein positron emission tomography, and imaging mass cytometry revealed a shift from resting to activated microglia. In summary, we prove a role for microglia in ICANS pathophysiology, identify the TAK1-NF-κB-p38 MAPK axis as a pathogenic signaling pathway and provide a rationale to test TAK1 inhibition in a clinical trial for ICANS prevention after CAR19 T cell-based cancer immunotherapy.


Sujet(s)
MAP Kinase Kinase Kinases , Microglie , Syndromes neurotoxiques , Récepteurs chimériques pour l'antigène , Animaux , Souris , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Microglie/immunologie , Microglie/métabolisme , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/immunologie , Humains , Récepteurs chimériques pour l'antigène/immunologie , Immunothérapie adoptive/méthodes , p38 Mitogen-Activated Protein Kinases/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Lymphome B/immunologie , Antigènes CD19/immunologie , Femelle , Lymphocytes T/immunologie , Transduction du signal
11.
Nat Commun ; 15(1): 4326, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773113

RÉSUMÉ

Resolving inflammation is thought to return the affected tissue back to homoeostasis but recent evidence supports a non-linear model of resolution involving a phase of prolonged immune activity. Here we show that within days following resolution of Streptococcus pneumoniae-triggered lung inflammation, there is an influx of antigen specific lymphocytes with a memory and tissue-resident phenotype as well as macrophages bearing alveolar or interstitial phenotype. The transcriptome of these macrophages shows enrichment of genes associated with prostaglandin biosynthesis and genes that drive T cell chemotaxis and differentiation. Therapeutic depletion of post-resolution macrophages, inhibition of prostaglandin E2 (PGE2) synthesis or treatment with an EP4 antagonist, MF498, reduce numbers of lung CD4+/CD44+/CD62L+ and CD4+/CD44+/CD62L-/CD27+ T cells as well as their expression of the α-integrin, CD103. The T cells fail to reappear and reactivate upon secondary challenge for up to six weeks following primary infection. Concomitantly, EP4 antagonism through MF498 causes accumulation of lung macrophages and marked tissue fibrosis. Our study thus shows that PGE2 signalling, predominantly via EP4, plays an important role during the second wave of immune activity following resolution of inflammation. This secondary immune activation drives local tissue-resident T cell development while limiting tissue injury.


Sujet(s)
Macrophages , Pneumonie à pneumocoques , Streptococcus pneumoniae , Mâle , Souris , Dinoprostone/antagonistes et inhibiteurs , Dinoprostone/métabolisme , Fibrose , Inflammation/immunologie , Inflammation/anatomopathologie , Poumon/immunologie , Poumon/microbiologie , Poumon/anatomopathologie , Lymphocytes/cytologie , Lymphocytes/immunologie , Macrophages/cytologie , Macrophages/immunologie , Macrophages alvéolaires/cytologie , Macrophages alvéolaires/immunologie , Souris de lignée C57BL , Phagocytes/cytologie , Phagocytes/immunologie , Pneumonie à pneumocoques/immunologie , Pneumonie à pneumocoques/anatomopathologie , Prostaglandines/biosynthèse , Quinoléines/administration et posologie , Streptococcus pneumoniae/physiologie , Sulfonamides/administration et posologie , Lymphocytes T/cytologie , Lymphocytes T/immunologie , Transcriptome , Animaux
12.
Haematologica ; 109(8): 2487-2499, 2024 08 01.
Article de Anglais | MEDLINE | ID: mdl-38572559

RÉSUMÉ

Innate myeloid cells especially neutrophils and their extracellular traps are known to promote intravascular coagulation and thrombosis formation in infections and various other conditions. Innate myeloid cell-dependent fibrin formation can support systemic immunity while its dysregulation enhances the severity of infectious diseases. Less is known about the immune mechanisms preventing dysregulation of fibrin homeostasis in infection. During experimental systemic infections local fibrin deposits in the liver microcirculation cause rapid arrest of CD4+ T cells. Arrested T-helper cells mostly represent Th17 cells that partially originate from the small intestine. Intravascular fibrin deposits activate mouse and human CD4+ T cells which can be mediated by direct fibrin-CD4+ T-cell interactions. Activated CD4+ T cells suppress fibrin deposition and microvascular thrombosis by directly counteracting coagulation activation by neutrophils and classical monocytes. T-cell activation, which is initially triggered by IL-12p40- and MHC-II-dependent mechanisms, enhances intravascular fibrinolysis via LFA-1. Moreover, CD4+ T cells disfavor the association of the thrombin-activatable fibrinolysis inhibitor (TAFI) with fibrin whereby fibrin deposition is increased by TAFI in the absence but not in the presence of T cells. In human infections thrombosis development is inversely related to microvascular levels of CD4+ T cells. Thus, fibrin promotes LFA-1-dependent T-helper cell activation in infections which drives a negative feedback cycle that rapidly restricts intravascular fibrin and thrombosis development.


Sujet(s)
Lymphocytes T CD4+ , Fibrine , Humains , Fibrine/métabolisme , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Infections/immunologie , Activation des lymphocytes/immunologie , Thrombose/étiologie , Thrombose/immunologie
13.
Brain Behav Immun ; 119: 572-596, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38663771

RÉSUMÉ

Chronic stress enhances the risk of neuropsychiatric disorders and contributes to the aggravation and chronicity of pain. The development of stress-associated diseases, including pain, is affected by individual vulnerability or resilience to stress, although the mechanisms remain elusive. We used the repeated social defeat stress model promoting susceptible and resilient phenotypes in male and female mice and induced knee mono-arthritis to investigate the impact of stress vulnerability on pain and immune system regulation. We analyzed different pain-related behaviors, measured blood cytokine and immune cell levels, and performed histological analyses at the knee joints and pain/stress-related brain areas. Stress susceptible male and female mice showed prolonged arthritis-associated hypersensitivity. Interestingly, hypersensitivity was exacerbated in male but not female mice. In males, stress promoted transiently increased neutrophils and Ly6Chigh monocytes, lasting longer in susceptible than resilient mice. While resilient male mice displayed persistently increased levels of the anti-inflammatory interleukin (IL)-10, susceptible mice showed increased levels of the pro-inflammatory IL-6 at the early- and IL-12 at the late arthritis stage. Although joint inflammation levels were comparable among groups, macrophage and neutrophil infiltration was higher in the synovium of susceptible mice. Notably, only susceptible male mice, but not females, presented microgliosis and monocyte infiltration in the prefrontal cortex at the late arthritis stage. Blood Ly6Chigh monocyte depletion during the early inflammatory phase abrogated late-stage hypersensitivity and the associated histological alterations in susceptible male mice. Thus, recruitment of blood Ly6Chigh monocytes during the early arthritis phase might be a key factor mediating the persistence of arthritis pain in susceptible male mice. Alternative neuro-immune pathways that remain to be explored might be involved in females.


Sujet(s)
Défaite sociale , Stress psychologique , Animaux , Mâle , Femelle , Souris , Stress psychologique/complications , Stress psychologique/immunologie , Stress psychologique/métabolisme , Souris de lignée C57BL , Cytokines/métabolisme , Arthrite/immunologie , Arthrite/métabolisme , Arthrite expérimentale/immunologie , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Cortex préfrontal/métabolisme , Hypersensibilité/immunologie , Hypersensibilité/métabolisme , Inflammation/métabolisme , Inflammation/immunologie , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/immunologie , Douleur/métabolisme , Monocytes/métabolisme , Monocytes/immunologie , Encéphale/métabolisme , Encéphale/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Modèles animaux de maladie humaine , Facteurs sexuels
14.
Blood ; 144(2): 216-226, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38648571

RÉSUMÉ

ABSTRACT: Triple-negative breast cancer (TNBC) is an aggressive tumor entity in which immune checkpoint (IC) molecules are primarily synthesized in the tumor environment. Here, we report that procoagulant platelets bear large amounts of such immunomodulatory factors and that the presence of these cellular blood components in TNBC relates to protumorigenic immune-cell activity and impaired survival. Mechanistically, tumor-released nucleic acids attract platelets to the aberrant tumor microvasculature, where they undergo procoagulant activation, thus delivering specific stimulatory and inhibitory IC molecules. This concomitantly promotes protumorigenic myeloid leukocyte responses and compromises antitumorigenic lymphocyte activity, ultimately supporting tumor growth. Interference with platelet-leukocyte interactions prevented immune cell misguidance and suppressed tumor progression, nearly as effective as systemic IC inhibition. Hence, our data uncover a self-sustaining mechanism of TNBC by using platelets to misdirect immune-cell responses. Targeting this irregular multicellular interplay may represent a novel immunotherapeutic strategy for TNBC without the adverse effects of systemic IC inhibition.


Sujet(s)
Plaquettes , Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/anatomopathologie , Humains , Plaquettes/immunologie , Plaquettes/anatomopathologie , Plaquettes/métabolisme , Femelle , Souris , Animaux , Échappement de la tumeur à la surveillance immunitaire , Lignée cellulaire tumorale , Échappement immunitaire
15.
Environ Microbiol Rep ; 16(2): e13266, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38653477

RÉSUMÉ

The Gram-positive bacteria Streptomyces davaonensis and Streptomyces cinnabarinus have been the only organisms known to produce roseoflavin, a riboflavin (vitamin B2) derived red antibiotic. Using a selective growth medium and a phenotypic screening, we were able to isolate a novel roseoflavin producer from a German soil sample. The isolation procedure was repeated twice, that is, the same strain could be isolated from the same location in Berlin 6 months and 12 months after its first isolation. Whole genome sequencing of the novel roseoflavin producer revealed an unusual chromosomal arrangement and the deposited genome sequence of the new isolate (G + C content of 71.47%) contains 897 genes per inverted terminal repeat, 6190 genes in the core and 107 genes located on an illegitimate terminal end. We identified the roseoflavin biosynthetic genes rosA, rosB and rosC and an unusually high number of riboflavin biosynthetic genes. Overexpression of rosA, rosB and rosC in Escherichia coli and enzyme assays confirmed their predicted functions in roseoflavin biosynthesis. A full taxonomic analysis revealed that the isolate represents a previously unknown Streptomyces species and we propose the name Streptomyces berlinensis sp. nov. for this roseoflavin producer.


Sujet(s)
Phylogenèse , Riboflavine , Riboflavine/analogues et dérivés , Microbiologie du sol , Streptomyces , Streptomyces/génétique , Streptomyces/classification , Streptomyces/métabolisme , Streptomyces/isolement et purification , Riboflavine/métabolisme , Riboflavine/biosynthèse , Composition en bases nucléiques , Génome bactérien , Séquençage du génome entier , Allemagne , Antibactériens/biosynthèse , Antibactériens/métabolisme
16.
Nat Commun ; 15(1): 2788, 2024 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-38555356

RÉSUMÉ

Hospital-acquired pneumonia (HAP) is associated with high mortality and costs, and frequently caused by multidrug-resistant (MDR) bacteria. Although prior antimicrobial therapy is a major risk factor for HAP, the underlying mechanism remains incompletely understood. Here, we demonstrate that antibiotic therapy in hospitalized patients is associated with decreased diversity of the gut microbiome and depletion of short-chain fatty acid (SCFA) producers. Infection experiments with mice transplanted with patient fecal material reveal that these antibiotic-induced microbiota perturbations impair pulmonary defense against MDR Klebsiella pneumoniae. This is dependent on inflammatory monocytes (IMs), whose fatty acid receptor (FFAR)2/3-controlled and phagolysosome-dependent antibacterial activity is compromized in mice transplanted with antibiotic-associated patient microbiota. Collectively, we characterize how clinically relevant antibiotics affect antimicrobial defense in the context of human microbiota, and reveal a critical impairment of IM´s antimicrobial activity. Our study provides additional arguments for the rational use of antibiotics and offers mechanistic insights for the development of novel prophylactic strategies to protect high-risk patients from HAP.


Sujet(s)
Antibactériens , Anti-infectieux , Humains , Souris , Animaux , Antibactériens/pharmacologie , Antibactériens/usage thérapeutique , Monocytes , Anti-infectieux/pharmacologie , Klebsiella pneumoniae , Poumon
17.
Clin Cancer Res ; 30(9): 1945-1958, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38427437

RÉSUMÉ

PURPOSE: Radiotherapy (RT) is a widely employed anticancer treatment. Emerging evidence suggests that RT can elicit both tumor-inhibiting and tumor-promoting immune effects. The purpose of this study is to investigate immune suppressive factors of radiotherapy. EXPERIMENTAL DESIGN: We used a heterologous two-tumor model in which adaptive concomitant immunity was eliminated. RESULTS: Through analysis of PD-L1 expression and myeloid-derived suppressor cells (MDSC) frequencies using patient peripheral blood mononuclear cells and murine two-tumor and metastasis models, we report that local irradiation can induce a systemic increase in MDSC, as well as PD-L1 expression on dendritic cells and myeloid cells, and thereby increase the potential for metastatic dissemination in distal, nonirradiated tissue. In a mouse model using two distinct tumors, we found that PD-L1 induction by ionizing radiation was dependent on elevated chemokine CXCL10 signaling. Inhibiting PD-L1 or MDSC can potentially abrogate RT-induced metastasis and improve clinical outcomes for patients receiving RT. CONCLUSIONS: Blockade of PD-L1/CXCL10 axis or MDSC infiltration during irradiation can enhance abscopal tumor control and reduce metastasis.


Sujet(s)
Antigène CD274 , Cellules myéloïdes suppressives , Animaux , Antigène CD274/métabolisme , Souris , Cellules myéloïdes suppressives/immunologie , Cellules myéloïdes suppressives/métabolisme , Humains , Métastase tumorale , Lignée cellulaire tumorale , Femelle , Modèles animaux de maladie humaine , Chimiokine CXCL10/métabolisme
19.
Circulation ; 149(1): 48-66, 2024 01 02.
Article de Anglais | MEDLINE | ID: mdl-37746718

RÉSUMÉ

BACKGROUND: Immune checkpoint inhibitors (ICIs), antibodies targeting PD-1 (programmed cell death protein 1)/PD-L1 (programmed death-ligand 1) or CTLA4 (cytotoxic T-lymphocyte-associated protein 4), have revolutionized cancer management but are associated with devastating immune-related adverse events including myocarditis. The main risk factor for ICI myocarditis is the use of combination PD-1 and CTLA4 inhibition. ICI myocarditis is often fulminant and is pathologically characterized by myocardial infiltration of T lymphocytes and macrophages. Although much has been learned about the role of T-cells in ICI myocarditis, little is understood about the identity, transcriptional diversity, and functions of infiltrating macrophages. METHODS: We used an established murine ICI myocarditis model (Ctla4+/-Pdcd1-/- mice) to explore the cardiac immune landscape using single-cell RNA-sequencing, immunostaining, flow cytometry, in situ RNA hybridization, molecular imaging, and antibody neutralization studies. RESULTS: We observed marked increases in CCR2 (C-C chemokine receptor type 2)+ monocyte-derived macrophages and CD8+ T-cells in this model. The macrophage compartment was heterogeneous and displayed marked enrichment in an inflammatory CCR2+ subpopulation highly expressing Cxcl9 (chemokine [C-X-C motif] ligand 9), Cxcl10 (chemokine [C-X-C motif] ligand 10), Gbp2b (interferon-induced guanylate-binding protein 2b), and Fcgr4 (Fc receptor, IgG, low affinity IV) that originated from CCR2+ monocytes. It is important that a similar macrophage population expressing CXCL9, CXCL10, and CD16α (human homologue of mouse FcgR4) was expanded in patients with ICI myocarditis. In silico prediction of cell-cell communication suggested interactions between T-cells and Cxcl9+Cxcl10+ macrophages via IFN-γ (interferon gamma) and CXCR3 (CXC chemokine receptor 3) signaling pathways. Depleting CD8+ T-cells or macrophages and blockade of IFN-γ signaling blunted the expansion of Cxcl9+Cxcl10+ macrophages in the heart and attenuated myocarditis, suggesting that this interaction was necessary for disease pathogenesis. CONCLUSIONS: These data demonstrate that ICI myocarditis is associated with the expansion of a specific population of IFN-γ-induced inflammatory macrophages and suggest the possibility that IFN-γ blockade may be considered as a treatment option for this devastating condition.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Myocardite , Humains , Souris , Animaux , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Lymphocytes T CD8+ , Myocardite/induit chimiquement , Myocardite/métabolisme , Récepteur-1 de mort cellulaire programmée , Antigène CTLA-4 , Ligands , Chimiokines/métabolisme , Macrophages/métabolisme , ARN/métabolisme
20.
Front Immunol ; 14: 1267866, 2023.
Article de Anglais | MEDLINE | ID: mdl-37849753

RÉSUMÉ

Tumor development and progression is shaped by the tumor microenvironment (TME), a heterogeneous assembly of infiltrating and resident host cells, their secreted mediators and intercellular matrix. In this context, tumors are infiltrated by various immune cells with either pro-tumoral or anti-tumoral functions. Recently, we published our non-invasive immunization platform DIVA suitable as a therapeutic vaccination method, further optimized by repeated application (DIVA2). In our present work, we revealed the therapeutic effect of DIVA2 in an MC38 tumor model and specifically focused on the mechanisms induced in the TME after immunization. DIVA2 resulted in transient tumor control followed by an immune evasion phase within three weeks after the initial tumor inoculation. High-dimensional flow cytometry analysis and single-cell mRNA-sequencing of tumor-infiltrating leukocytes revealed cytotoxic CD8+ T cells as key players in the immune control phase. In the immune evasion phase, inflammatory CCR2+ PDL-1+ monocytes with immunosuppressive properties were recruited into the tumor leading to suppression of DIVA2-induced tumor-reactive T cells. Depletion of CCR2+ cells with specific antibodies resulted in prolonged survival revealing CCR2+ monocytes as important for tumor immune escape in the TME. In summary, the present work provides a platform for generating a strong antigen-specific primary and memory T cell immune response using the optimized transcutaneous immunization method DIVA2. This enables protection against tumors by therapeutic immune control of solid tumors and highlights the immunosuppressive influence of tumor infiltrating CCR2+ monocytes that need to be inactivated in addition for successful cancer immunotherapy.


Sujet(s)
Monocytes , Tumeurs , Humains , Lymphocytes T CD8+ , Lymphocytes T cytotoxiques , Immunothérapie , Tumeurs/thérapie , Microenvironnement tumoral , Récepteurs CCR2
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE