Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Nat Immunol ; 25(5): 802-819, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38684922

RESUMEN

Sepsis induces immune alterations, which last for months after the resolution of illness. The effect of this immunological reprogramming on the risk of developing cancer is unclear. Here we use a national claims database to show that sepsis survivors had a lower cumulative incidence of cancers than matched nonsevere infection survivors. We identify a chemokine network released from sepsis-trained resident macrophages that triggers tissue residency of T cells via CCR2 and CXCR6 stimulations as the immune mechanism responsible for this decreased risk of de novo tumor development after sepsis cure. While nonseptic inflammation does not provoke this network, laminarin injection could therapeutically reproduce the protective sepsis effect. This chemokine network and CXCR6 tissue-resident T cell accumulation were detected in humans with sepsis and were associated with prolonged survival in humans with cancer. These findings identify a therapeutically relevant antitumor consequence of sepsis-induced trained immunity.


Asunto(s)
Macrófagos , Neoplasias , Sepsis , Humanos , Sepsis/inmunología , Macrófagos/inmunología , Femenino , Neoplasias/inmunología , Neoplasias/terapia , Masculino , Receptores CXCR6/metabolismo , Animales , Linfocitos T/inmunología , Receptores CCR2/metabolismo , Persona de Mediana Edad , Ratones , Anciano , Quimiocinas/metabolismo , Adulto
2.
Nat Commun ; 14(1): 4290, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37463962

RESUMEN

Endo-lysosomes transport along microtubules and clustering in the perinuclear area are two necessary steps for microbes to activate specialized phagocyte functions. We report that RUN and FYVE domain-containing protein 3 (RUFY3) exists as two alternative isoforms distinguishable by the presence of a C-terminal FYVE domain and by their affinity for phosphatidylinositol 3-phosphate on endosomal membranes. The FYVE domain-bearing isoform (iRUFY3) is preferentially expressed in primary immune cells and up-regulated upon activation by microbes and Interferons. iRUFY3 is necessary for ARL8b + /LAMP1+ endo-lysosomes positioning in the pericentriolar organelles cloud of LPS-activated macrophages. We show that iRUFY3 controls macrophages migration, MHC II presentation and responses to Interferon-γ, while being important for intracellular Salmonella replication. Specific inactivation of rufy3 in phagocytes leads to aggravated pathologies in mouse upon LPS injection or bacterial pneumonia. This study highlights the role of iRUFY3 in controlling endo-lysosomal dynamics, which contributes to phagocyte activation and immune response regulation.


Asunto(s)
Presentación de Antígeno , Lipopolisacáridos , Animales , Ratones , Endosomas/metabolismo , Lipopolisacáridos/metabolismo , Lisosomas/metabolismo , Fagocitos
3.
Am J Respir Crit Care Med ; 206(3): 295-310, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35486851

RESUMEN

Rationale: Brain injury induces systemic immunosuppression, increasing the risk of viral reactivations and altering neurological recovery. Objectives: To determine if systemic immune alterations and lung replication of herpesviridae are associated and can help predict outcomes after brain injury. Methods: We collected peripheral blood mononuclear cells in patients with severe brain injury requiring invasive mechanical ventilation. We systematically searched for respiratory herpes simplex virus (HSV) replications in tracheal aspirates. We also performed chromatin immunoprecipitation sequencing, RNA-sequencing, and in vitro functional assays of monocytes and CD4 T cells collected on Day 1 to characterize the immune response to severe acute brain injury. The primary outcome was the Glasgow Outcome Scale Extended at 6 months. Measurements and Main Results: In 344 patients with severe brain injury, lung HSV reactivations were observed in 39% of the 232 patients seropositive for HSV and independently associated with poor neurological recovery at 6 months (hazard ratio, 1.90; 95% confidence interval, 1.08-3.57). Weighted gene coexpression network analyses of the transcriptomic response of monocytes to brain injury defined a module of 721 genes, including PD-L1 and CD80, enriched for the binding DNA motif of the transcriptional factor Zeb2 and whose ontogenic analyses revealed decreased IFN-γ-mediated and antiviral response signaling pathways. This monocyte signature was preserved in a validation cohort and predicted the neurological outcome at 6 months with good accuracy (area under the curve, 0.786; 95% confidence interval, 0.593-0.978). Conclusions: A specific monocyte signature is associated with HSV reactivation and predicts poor recovery after brain injury. The alterations of the immune control of herpesviridae replication are understudied and represent a novel therapeutic target.


Asunto(s)
Lesiones Encefálicas , Herpes Simple , Herpesvirus Humano 1 , Herpesvirus Humano 1/genética , Humanos , Leucocitos Mononucleares , Monocitos
4.
Cells ; 10(10)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34685700

RESUMEN

At the early stages of life development, alveoli are colonized by embryonic macrophages, which become resident alveolar macrophages (ResAM) and self-sustain by local division. Genetic and epigenetic signatures and, to some extent, the functions of ResAM are dictated by the lung microenvironment, which uses cytokines, ligand-receptor interactions, and stroma cells to orchestrate lung homeostasis. In resting conditions, the lung microenvironment induces in ResAM a tolerogenic programming that prevents unnecessary and potentially harmful inflammation responses to the foreign bodies, which continuously challenge the airways. Throughout life, any episode of acute inflammation, pneumonia being likely the most frequent cause, depletes the pool of ResAM, leaving space for the recruitment of inflammatory monocytes that locally develop in monocyte-derived alveolar macrophages (InfAM). During lung infection, the local microenvironment induces a temporary inflammatory signature to the recruited InfAM to handle the tissue injury and eliminate the pathogens. After a few days, the recruited InfAM, which locally self-sustain and develop as new ResAM, gain profibrotic functions required for tissue healing. After the complete resolution of the infectious episode, the functional programming of both embryonic and monocyte-derived ResAM remains altered for months and possibly for the entire life. Adult lungs thus contain a wide diversity of ResAM since every infection brings new waves of InfAM which fill the room left open by the inflammatory process. The memory of these innate cells called trained immunity constitutes an immunologic scar left by inflammation, notably pneumonia. This memory of ResAM has advantages and drawbacks. In some cases, lung-trained immunity offers better defense capacities against autoimmune disorders and the long-term risk of infection. At the opposite, it can perpetuate a harmful process and lead to a pathological state, as is the case among critically ill patients who have immune paralysis and are highly susceptible to hospital-acquired pneumonia and acute respiratory distress syndrome. The progress in understanding the kinetics of response of alveolar macrophages (AM) to lung inflammation is paving the way to new treatments of pneumonia and lung inflammatory process.


Asunto(s)
Adaptación Fisiológica , Inflamación/patología , Macrófagos Alveolares/patología , Fibrosis , Homeostasis , Humanos , Infecciones/inmunología , Infecciones/patología , Inflamación/inmunología , Macrófagos Alveolares/inmunología
5.
Antimicrob Agents Chemother ; 65(12): e0076821, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34606337

RESUMEN

Staphylococcus aureus bone infections remain a therapeutic challenge, leading to long and expensive hospitalizations. Systemic antibiotic treatments are inconsistently effective, due to insufficient penetration into the infectious site. In an osteomyelitis model, the single local administration of nanoparticle-encapsulated daptomycin allows sterilization of the infectious sites after 4 and 14 days of treatment, while daily systemic daptomycin treatment for 4 days was not effective. These results demonstrate the great potential of this local antibiotic treatment.


Asunto(s)
Artritis Infecciosa , Daptomicina , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Antibacterianos/uso terapéutico , Artritis Infecciosa/tratamiento farmacológico , Daptomicina/uso terapéutico , Humanos , Infecciones Estafilocócicas/tratamiento farmacológico
6.
Int J Mol Sci ; 22(6)2021 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-33806791

RESUMEN

The world is on the verge of a major antibiotic crisis as the emergence of resistant bacteria is increasing, and very few novel molecules have been discovered since the 1960s. In this context, scientists have been exploring alternatives to conventional antibiotics, such as ribosomally synthesized and post-translationally modified peptides (RiPPs). Interestingly, the highly potent in vitro antibacterial activity and safety of ruminococcin C1, a recently discovered RiPP belonging to the sactipeptide subclass, has been demonstrated. The present results show that ruminococcin C1 is efficient at curing infection and at protecting challenged mice from Clostridium perfringens with a lower dose than the conventional antibiotic vancomycin. Moreover, antimicrobial peptide (AMP) is also effective against this pathogen in the complex microbial community of the gut environment, with a selective impact on a few bacterial genera, while maintaining a global homeostasis of the microbiome. In addition, ruminococcin C1 exhibits other biological activities that could be beneficial for human health, as well as other fields of applications. Overall, this study, by using an in vivo infection approach, confirms the antimicrobial clinical potential and highlights the multiple functional properties of ruminococcin C1, thus extending its therapeutic interest.


Asunto(s)
Antibacterianos/farmacología , Bacteriocinas/farmacología , Péptidos/farmacología , Antibacterianos/química , Antifúngicos/farmacología , Bacteriocinas/química , Biopelículas/efectos de los fármacos , Clostridiales/metabolismo , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/microbiología , Clostridium perfringens/efectos de los fármacos , Humanos , Péptidos/química , Procesamiento Proteico-Postraduccional
7.
Infect Immun ; 88(12)2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32928966

RESUMEN

Natural killer (NK) cells play a key role in both antibacterial and antitumor immunity. Pseudomonas aeruginosa infection has already been reported to alter NK cell functions. We studied in vitro the effect of P. aeruginosa on NK cell cytotoxic response (CD107a membrane expression) to a lymphoma cell line. Through positive and negative cell sorting and adoptive transfer, we determined the influence of monocytes, lymphocytes, and regulatory T cells (Treg) on NK cell function during P. aeruginosa infection. We also studied the role of the activating receptor natural killer group 2D (NKG2D) in NK cell response to B221. We determined that P. aeruginosa significantly altered both cytotoxic response to B221 and NKG2D expression on NK cells in a Treg-dependent manner and that the NKG2D receptor was involved in NK cell cytotoxic response to B221. Our results also suggested that during P. aeruginosa infection, monocytes participated in Treg-mediated NK cell alteration. In conclusion, P. aeruginosa infection impairs NK cell cytotoxicity and alters antitumor immunity. These results highlight the strong interaction between bacterial infection and immunity against cancer.


Asunto(s)
Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Linfocitos T Reguladores/inmunología , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Pruebas Inmunológicas de Citotoxicidad , Humanos , Leucocitos Mononucleares , Receptores de Lipopolisacáridos/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Monocitos/inmunología , Infecciones por Pseudomonas/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo
8.
J Infect Dis ; 222(7): 1222-1234, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32697326

RESUMEN

Sepsis causes inflammation-induced immunosuppression with lymphopenia and alterations of CD4+ T-cell functions that renders the host prone to secondary infections. Whether and how regulatory T cells (Treg) are involved in this postseptic immunosuppression is unknown. We observed in vivo that early activation of Treg during Staphylococcus aureus sepsis induces CD4+ T-cell impairment and increases susceptibility to secondary pneumonia. The tumor necrosis factor receptor 2 positive (TNFR2pos) Treg subset endorsed the majority of effector immunosuppressive functions, and TNRF2 was particularly associated with activation of genes involved in cell cycle and replication in Treg, probably explaining their maintenance. Blocking or deleting TNFR2 during sepsis decreased the susceptibility to secondary infection. In humans, our data paralleled those in mice; the expression of CTLA-4 was dramatically increased in TNFR2pos Treg after culture in vitro with S. aureus. Our findings describe in vivo mechanisms underlying sepsis-induced immunosuppression and identify TNFR2pos Treg as targets for therapeutic intervention.


Asunto(s)
Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Sepsis/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Células Cultivadas , Femenino , Humanos , Terapia de Inmunosupresión , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Sepsis/microbiología , Staphylococcus aureus , Linfocitos T Reguladores/citología
10.
Nat Immunol ; 21(6): 636-648, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32424365

RESUMEN

Sepsis and trauma cause inflammation and elevated susceptibility to hospital-acquired pneumonia. As phagocytosis by macrophages plays a critical role in the control of bacteria, we investigated the phagocytic activity of macrophages after resolution of inflammation. After resolution of primary pneumonia, murine alveolar macrophages (AMs) exhibited poor phagocytic capacity for several weeks. These paralyzed AMs developed from resident AMs that underwent an epigenetic program of tolerogenic training. Such adaptation was not induced by direct encounter of the pathogen but by secondary immunosuppressive signals established locally upon resolution of primary infection. Signal-regulatory protein α (SIRPα) played a critical role in the establishment of the microenvironment that induced tolerogenic training. In humans with systemic inflammation, AMs and also circulating monocytes still displayed alterations consistent with reprogramming six months after resolution of inflammation. Antibody blockade of SIRPα restored phagocytosis in monocytes of critically ill patients in vitro, which suggests a potential strategy to prevent hospital-acquired pneumonia.


Asunto(s)
Epigénesis Genética , Inflamación/etiología , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Animales , Biomarcadores , Reprogramación Celular , Citocinas/metabolismo , Humanos , Tolerancia Inmunológica , Inmunofenotipificación , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Pulmón/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos Alveolares/inmunología , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Fagocitosis/inmunología , Neumonía/etiología , Neumonía/metabolismo , Neumonía/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
11.
Mol Immunol ; 118: 52-59, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31855807

RESUMEN

BACKGROUND: Interleukin (IL)-22 is a cytokine involved in tissue protection and repair following lung pathologies. Interferon (IFN)-λ cytokines displayed similar properties during viral infection and a synergy of action between these two players has been documented in the intestine. We hypothesize that during Pseudomonas aeruginosa challenge, IL-22 up-regulates IFN-λ and that IFN-λ exhibits protective functions during Pseudomonas aeruginosa acute pneumonia model in mice. METHODS: Using an in vitro human alveolar epithelial cell line A549, we assessed the ability of IL-22 to enhance IFN-λ expression during infection. IFN-λ protective function was evaluated in an acute mouse pneumonia model. RESULTS: We first demonstrated in murine lungs that only type-II alveolar cells express IL-22 receptor and that IL-22 treatment of A549 cell line up-regulates IFN-λ expression. In a murine acute pneumonia model, IL-22 administration maintained significant IFN-λ levels in the broncho-alveolar fluids whereas IL-22 neutralization abolished IFN-λ up-regulation. In vivo administration of IFN-λ during Pseudomonas aeruginosa pneumonia improves mice outcome by dampening neutrophil recruitment and decreasing epithelium damages. DISCUSSION: We show here that IL-22 regulates IFN-λ levels during Pseudomonas aeruginosa pneumonia.


Asunto(s)
Interferones/inmunología , Interleucinas/inmunología , Neumonía/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Células A549 , Células Epiteliales Alveolares/inmunología , Animales , Bronquios/inmunología , Línea Celular Tumoral , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/inmunología , Ratones , Infiltración Neutrófila/inmunología , Receptores de Interleucina/inmunología , Regulación hacia Arriba/inmunología , Interleucina-22
12.
Artículo en Inglés | MEDLINE | ID: mdl-30858223

RESUMEN

Impeding, as well as reducing, the burden of antimicrobial resistance in Gram-negative pathogens is an urgent public health endeavor. Our current antibiotic armamentarium is dwindling, while major resistance determinants (e.g., extended-spectrum ß-lactamases [ESBLs]) continue to evolve and disseminate around the world. One approach to attack this problem is to develop novel therapies that will protect our current agents. AAI101 is a novel penicillanic acid sulfone ß-lactamase inhibitor similar in structure to tazobactam, with one important difference. AAI101 possesses a strategically placed methyl group that gives the inhibitor a net neutral charge, enhancing bacterial cell penetration. AAI101 paired with cefepime, also a zwitterion, is in phase III of clinical development for the treatment of serious Gram-negative infections. Here, AAI101 was found to restore the activity of cefepime against class A ESBLs (e.g., CTX-M-15) and demonstrated increased potency compared to that of piperacillin-tazobactam when tested against an established isogenic panel. The enzymological properties of AAI101 further revealed that AAI101 possessed a unique mechanism of ß-lactamase inhibition compared to that of tazobactam. Additionally, upon reaction with AAI101, CTX-M-15 was modified to an inactive state. Notably, the in vivo efficacy of cefepime-AAI101 was demonstrated using a mouse septicemia model, indicating the ability of AAI101 to bolster significantly the therapeutic efficacy of cefepime in vivo The combination of AAI101 with cefepime represents a potential carbapenem-sparing treatment regimen for infections suspected to be caused by Enterobacteriaceae expressing ESBLs.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Cefepima/farmacología , Enterobacteriaceae/efectos de los fármacos , Enterobacteriaceae/metabolismo , Combinación Piperacilina y Tazobactam/farmacología , Sulbactam/farmacología , Triazoles/farmacología , Inhibidores de beta-Lactamasas/farmacología , Espectrometría de Masa por Ionización de Electrospray
13.
Crit Care Med ; 47(1): e28-e35, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30303841

RESUMEN

OBJECTIVES: In patients with spinal cord injury, spinal cord injury-immune depression syndrome induces pneumonia. We aimed to develop a new spinal cord injury-immune depression syndrome mouse model and to test antiprogrammed cell death 1 therapy. DESIGN: Experimental study. SETTING: Research laboratory. SUBJECTS: RjOrl: SWISS and BALB/cJ mice. INTERVENTIONS: Mouse model of spinal cord injury-immune depression syndrome followed by a methicillin-susceptible Staphylococcus aureus pneumonia. Lung injuries were assessed by histologic analysis. Membrane markers and intracytoplasmic cytokines were assessed by flow cytometry. Cytokine production was assessed by quantitative polymerase chain reaction (messenger RNA) and enzyme-linked immunosorbent assay (protein). Animals were treated with blocking antiprogrammed cell death 1 antibodies (intraperitoneal injection). MEASUREMENTS AND MAIN RESULTS: Spinal cord injury mice were more susceptible to methicillin-susceptible S. aureus pneumonia (increased mortality rate). An early inflammatory response was observed in spinal cord injury mice characterized in lungs by a decreased percentage of aerated tissue, an increased production of proinflammatory cytokines (tumor necrosis factor-α). In spleen, an increased expression of major histocompatibility complex class II molecules on dendritic cells, and an increased production of proinflammatory cytokines (interleukin-12, interferon-γ) was observed. Following this pulmonary and systemic inflammation, spinal cord injury-immune depression syndrome was observed in spleens as acknowledged by a decrease of spleen's weight, a lymphopenia, a decrease of major histocompatibility complex class II expression on dendritic cells. An increase of interleukin-10 production and the increase of a cell exhaustion marker expression, programmed cell death 1 receptor on T-cell were also observed. Blockade of programmed cell death 1 molecules, improved survival of spinal cord injury infected mice and enhanced interferon-γ production by natural killer T cells as well as number of viable CD4 T cells. CONCLUSIONS: This model of spinal cord injury in mice mimics a clinical scenario rendering animals prone to a secondary pneumonia. We show for the first time an acute T-cell exhaustion-like phenomenon following an initial inflammatory response. Finally, inhibition of exhaustion pathway should be considered as a new therapeutic option to overcome spinal cord injury-immune depression syndrome and to decrease the rate of nosocomial pneumonia.


Asunto(s)
Anticuerpos/farmacología , Neumonía Bacteriana/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/inmunología , Traumatismos de la Médula Espinal/complicaciones , Staphylococcus aureus/inmunología , Animales , Citocinas/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Antígenos de Histocompatibilidad Clase II/inmunología , Ratones Endogámicos BALB C , Neumonía Bacteriana/microbiología , Bazo/metabolismo , Linfocitos T/inmunología
14.
J Antimicrob Chemother ; 73(9): 2418-2421, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29901764

RESUMEN

Objectives: To evaluate the significant role played by biofilms during prosthetic vascular material infections (PVMIs). Methods: We developed an in vivo mouse model of Staphylococcus aureus PVMI allowing its direct observation by confocal microscopy to describe: (i) the structure of biofilms developed on Dacron® vascular material; (ii) the localization and effect of antibiotics on these biostructures; and (iii) the interaction between bacteria and host tissues and cells during PVMI. Results: In this model we demonstrated that the biofilm structures are correlated to the activity of antibiotics. Furthermore, live S. aureus bacteria were visualized inside the macrophages present at the biofilm sites, which is significant as antibiotics do not penetrate these immune cells. Conclusions: This intracellular situation may explain the limited effect of antibiotics and also why PVMIs can relapse after antibiotic therapy.


Asunto(s)
Antibacterianos/uso terapéutico , Biopelículas/crecimiento & desarrollo , Citosol/microbiología , Macrófagos/microbiología , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/crecimiento & desarrollo , Animales , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Prótesis Vascular/efectos adversos , Prótesis Vascular/microbiología , Modelos Animales de Enfermedad , Femenino , Ratones , Microscopía Confocal , Infecciones Relacionadas con Prótesis/microbiología , Recurrencia , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Insuficiencia del Tratamiento
15.
Front Immunol ; 8: 1283, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29067027

RESUMEN

Pseudomonas aeruginosa (PA) expresses the type III secretion system (T3SS) and effector exoenzymes that interfere with intracellular pathways. Natural killer (NK) cells play a key role in antibacterial immunity and their activation is highly dependent on IL-12 produced by myeloid cells. We studied PA and NK cell interactions and the role of IL-12 using human peripheral blood mononuclear cells, sorted human NK cells, and a human NK cell line (NK92). We used a wild-type (WT) strain of PA (PAO1) or isogenic PA-deleted strains to delineate the role of T3SS and exoenzymes. Our hypotheses were tested in vivo in a PA-pneumonia mouse model. Human NK cells or NK92 cell line produced low levels of IFN-γ in response to PA without IL-12 stimulation, whereas PA significantly increased IFN-γ after IL-12 priming. The modulation of IFN-γ production by PA required bacteria-to-cell contact. Among T3SS effectors, exoenzyme T (ExoT) upregulates IFN-γ production and control ERK activation. In vivo, ExoT also increases IFN-γ levels and the percentage of IFN-γ+ NK cells in lungs during PA pneumonia, confirming in vitro data. In conclusion, our results suggest that T3SS could modulate the production of IFN-γ by NK cells after PA infection through ERK activation.

16.
Anaerobe ; 48: 172-176, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28859990

RESUMEN

Cutibacterium (formerly Propionibacterium) acnes is involved in chronic/low-grade pathologies such as sarcoidosis or prosthetic joint infection (PJI). In these diseases, granulomatous structures are frequently observed. In this study, we induced a physiological granulomatous reaction in response to different well-characterized clinical C. acnes isolates in order to investigate the cellular process during granuloma formation. Three C. acnes isolates selected according to their origin (PJI, sarcoidosis and acne) were typed by MLST. All C. acnes isolates generated granulomatous structures in our experimental conditions. The bacterial burden was better controlled by granulomas induced by the sarcoidosis C. acnes isolate. The PJI C. acnes isolate, belonging to CC36, promoted the recruitment of CD8+ lymphocytes inside the granuloma. In contrast, the acne and sarcoidosis C. acnes isolates, belonging to phylotypes IA1/CC18 and IA2/CC28, respectively, generated a higher number of granulomas and promoted the recruitment of CD4+ lymphocytes inside the granuloma. Our results provide new evidence supporting the role of C. acnes in the development of sarcoidosis and new explanations concerning the mechanisms underlying PJI due to C. acnes.


Asunto(s)
Infecciones por Bacterias Grampositivas/complicaciones , Infecciones por Bacterias Grampositivas/inmunología , Granuloma/etiología , Inmunidad , Propionibacterium acnes/inmunología , Susceptibilidad a Enfermedades , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/microbiología , Tipificación de Secuencias Multilocus , Propionibacterium acnes/clasificación , Propionibacterium acnes/genética
17.
Sci Rep ; 7(1): 11010, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28887540

RESUMEN

Pseudomonas aeruginosa is a major threat for immune-compromised patients. Bacterial pneumonia can induce uncontrolled and massive neutrophil recruitment ultimately leading to acute respiratory distress syndrome and epithelium damage. Interleukin-22 plays a central role in the protection of the epithelium. In this study, we aimed to evaluate the role of interleukin-22 and its soluble receptor IL-22BP in an acute Pseudomonas aeruginosa pneumonia model in mice. In this model, we noted a transient increase of IL-22 during Pseudomonas aeruginosa challenge. Using an antibody-based approach, we demonstrated that IL-22 neutralisation led to increased susceptibility to infection and to lung damage correlated with an increase in neutrophil accumulation in the lungs. On the contrary, rIL-22 administration or IL-22BP neutralisation led to a decrease in mouse susceptibility and lung damage associated with a decrease in neutrophil accumulation. This study demonstrated that the IL-22/IL-22BP system plays a major role during Pseudomonas aeruginosa pneumonia by moderating neutrophil accumulation in the lungs that ultimately leads to epithelium protection.


Asunto(s)
Interleucinas/análisis , Pulmón/patología , Infiltración Neutrófila , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/patología , Receptores de Interleucina/análisis , Animales , Modelos Animales de Enfermedad , Ratones , Interleucina-22
18.
J Glob Antimicrob Resist ; 10: 326-329, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28689923

RESUMEN

OBJECTIVES: Combination antibiotic therapy has been used successfully to treat some patients with bacterial infections. However, although certain combinations may result in beneficial synergistic activity, others may produce antagonistic effects resulting in poor treatment outcomes. Ceftolozane/tazobactam is an antibacterial agent with potent activity against Pseudomonas aeruginosa and many other Gram-negative pathogens, including extended-spectrum ß-lactamase (ESBL)-producing Enterobacteriaceae. This study aimed to evaluate potential synergistic or antagonistic interactions between ceftolozane/tazobactam and a selection of antibacterial agents. METHODS: Chequerboard analyses were conducted with Escherichia coli, Klebsiella pneumoniae and P. aeruginosa isolates. RESULTS: Combinations of ceftolozane/tazobactam with aztreonam, amikacin, tigecycline and meropenem resulted in synergistic effects in some of the bacterial strains tested. The potency of ceftolozane/tazobactam against common Gram-negative pathogens was not compromised in the presence of other commonly prescribed antibacterial agents, and ceftolozane/tazobactam did not antagonise the activity of these other antibacterials. CONCLUSIONS: The synergy observed for some antibacterial combinations in this study supplements the currently available information for combination therapy and may suggest new directions for treating challenging cases. Some synergistic effects may be attributed, at least in part, to the ESBL-inhibitory activity of tazobactam, although this remains to be proven. The mechanisms of the other synergistic interactions observed also require further elucidation. Ceftolozane/tazobactam did not adversely affect the activity of, and was not affected by, other antibacterial agents given concurrently. In vivo studies will be needed to substantiate these results and to determine their clinical relevance.


Asunto(s)
Cefalosporinas/farmacología , Quimioterapia Combinada/métodos , Bacterias Gramnegativas/efectos de los fármacos , Tazobactam/farmacología , Amicacina/farmacología , Aztreonam/farmacología , Antagonismo de Drogas , Sinergismo Farmacológico , Humanos , Meropenem/farmacología , Pruebas de Sensibilidad Microbiana , Tigeciclina/farmacología
19.
Immunity ; 47(1): 135-147.e5, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723546

RESUMEN

Lung infections cause prolonged immune alterations and elevated susceptibility to secondary pneumonia. We found that, after resolution of primary viral or bacterial pneumonia, dendritic cells (DC), and macrophages exhibited poor antigen-presentation capacity and secretion of immunogenic cytokines. Development of these "paralyzed" DCs and macrophages depended on the immunosuppressive microenvironment established upon resolution of primary infection, which involved regulatory T (Treg) cells and the cytokine TGF-ß. Paralyzed DCs secreted TGF-ß and induced local Treg cell accumulation. They also expressed lower amounts of IRF4, a transcription factor associated with increased antigen-presentation capacity, and higher amounts of Blimp1, a transcription factor associated with tolerogenic functions, than DCs present during primary infection. Blimp1 expression in DC of humans suffering sepsis or trauma correlated with severity and complicated outcomes. Our findings describe mechanisms underlying sepsis- and trauma-induced immunosuppression, reveal prognostic markers of susceptibility to secondary infections and identify potential targets for therapeutic intervention.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Escherichia coli/inmunología , Virus de la Influenza A/inmunología , Macrófagos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía/inmunología , Sepsis/inmunología , Anciano , Animales , Presentación de Antígeno , Diferenciación Celular , Células Cultivadas , Escherichia coli , Femenino , Humanos , Tolerancia Inmunológica , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Linfocitos T Reguladores/inmunología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
20.
Sci Rep ; 7: 42918, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28218305

RESUMEN

Cutibacterium acnes (formerly Propionibacterium acnes) is recognized as a pathogen in foreign-body infections (arthroplasty or spinal instrumentation). To date, the direct impact of C. acnes on bone cells has never been explored. The clade of 11 C. acnes clinical isolates was determined by MLST. Human osteoblasts and osteoclasts were infected by live C. acnes. The whole genome sequence of six isolates of this collection was analyzed. CC36 C. acnes strains were significantly less internalized by osteoblasts and osteoclasts than CC18 and CC28 C. acnes strains (p ≤ 0.05). The CC18 C. acnes ATCC6919 isolate could survive intracellularly for at least 96 hours. C. acnes significantly decreased the resorption ability of osteoclasts with a major impact by the CC36 strain (p ≤ 0.05). Genome analysis revealed 27 genes possibly linked to these phenotypic behaviors. We showed a direct impact of C. acnes on bone cells, providing new explanations about the development of C. acnes foreign-body infections.


Asunto(s)
Propionibacterium acnes/fisiología , Enfermedades Óseas/microbiología , Enfermedades Óseas/patología , Línea Celular Tumoral , Supervivencia Celular , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/patología , Humanos , Artropatías/microbiología , Artropatías/patología , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Tipificación de Secuencias Multilocus , Propionibacterium acnes/genética , Propionibacterium acnes/aislamiento & purificación , Secuenciación Completa del Genoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA