Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(44): 27608-27619, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33087560

RESUMEN

Streptococcus pneumoniae can cause disease in various human tissues and organs, including the ear, the brain, the blood, and the lung, and thus in highly diverse and dynamic environments. It is challenging to study how pneumococci control virulence factor expression, because cues of natural environments and the presence of an immune system are difficult to simulate in vitro. Here, we apply synthetic biology methods to reverse-engineer gene expression control in S. pneumoniae A selection platform is described that allows for straightforward identification of transcriptional regulatory elements out of combinatorial libraries. We present TetR- and LacI-regulated promoters that show expression ranges of four orders of magnitude. Based on these promoters, regulatory networks of higher complexity are assembled, such as logic AND gates and IMPLY gates. We demonstrate single-copy genome-integrated toggle switches that give rise to bimodal population distributions. The tools described here can be used to mimic complex expression patterns, such as the ones found for pneumococcal virulence factors. Indeed, we were able to rewire gene expression of the capsule operon, the main pneumococcal virulence factor, to be externally inducible (YES gate) or to act as an IMPLY gate (only expressed in absence of inducer). Importantly, we demonstrate that these synthetic gene-regulatory networks are functional in an influenza A virus superinfection murine model of pneumonia, paving the way for in vivo investigations of the importance of gene expression control on the pathogenicity of S. pneumoniae.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Infecciones Oportunistas/microbiología , Neumonía Neumocócica/microbiología , Neumonía Viral/virología , Streptococcus pneumoniae/patogenicidad , Sobreinfección/microbiología , Animales , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Redes Reguladoras de Genes , Genes Sintéticos/genética , Humanos , Virus de la Influenza A/patogenicidad , Masculino , Ratones , Nasofaringe/microbiología , Operón/genética , Infecciones Oportunistas/complicaciones , Neumonía Neumocócica/complicaciones , Neumonía Viral/complicaciones , Regiones Promotoras Genéticas/genética , Streptococcus pneumoniae/genética , Sobreinfección/complicaciones , Biología Sintética/métodos , Factores de Transcripción/metabolismo , Factores de Virulencia/metabolismo
2.
J Immunol ; 205(10): 2873-2882, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33008952

RESUMEN

The TLR5 agonist flagellin is a potent adjuvant and is currently being developed for use in vaccines. The mechanisms that drive flagellin's activity are influenced by its administration route. Previous studies showed that lung structural cells (especially epithelial cells lining the conducting airways) are pivotal for the efficacy of intranasally administered flagellin-containing vaccines. In this study, we looked at how the airway epithelial cells (AECs) regulate the flagellin-dependent stimulation of Ag-specific CD4+ T cells and the Ab response in mice. Our results demonstrate that after sensing flagellin, AECs trigger the release of GM-CSF in a TLR5-dependent fashion and the doubling of the number of activated type 2 conventional dendritic cells (cDC2s) in draining lymph nodes. Furthermore, the neutralization of GM-CSF reduced cDC2s activation. This resulted in lower of Ag-specific CD4+ T cell count and Ab titers in mice. Our data indicate that during pulmonary immunization, the GM-CSF released by AECs orchestrates the cross-talk between cDC2s and CD4+ T cells and thus drives flagellin's adjuvant effect.


Asunto(s)
Células Epiteliales/metabolismo , Flagelina/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Mucosa Respiratoria/inmunología , Vacunas/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Comunicación Celular/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Epiteliales/inmunología , Femenino , Flagelina/administración & dosificación , Inmunidad Mucosa , Inmunogenicidad Vacunal , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Noqueados , Modelos Animales , Cultivo Primario de Células , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Receptor Toll-Like 5/agonistas , Receptor Toll-Like 5/genética , Vacunas/administración & dosificación
3.
Eur J Immunol ; 49(12): 2134-2145, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31489613

RESUMEN

The 3-O-desacyl-4'-monophosphoryl lipid A (MPL) activates immunity through Toll-like receptor 4 (TLR4) signaling. The Adjuvant System AS01 contains MPL and is used in the candidate malaria vaccine and the licensed zoster vaccine. Recent studies reported that AS01 adjuvant activity depends on a transient inflammation at the site of vaccination, but the role of stromal or structural cells in the adjuvant effect is unknown. We investigated this question in mouse models by assessing the role of TLR4 on hematopoietic versus resident structural cells during immunization with AS01-adjuvanted vaccines. We first established that TLR4-deficient animals had a reduced immune response to an AS01-adjuvanted vaccine. Using bone marrow chimera, we consistently found that Tlr4 expression in radio-sensitive cells, i.e., hematopoietic cells, was required for an optimal adjuvant effect on antibody and T-cell responses. At day 1 after injection, the pro-inflammatory reaction at the site of injection was strongly dependent on TLR4 signaling in hematopoietic cells. Similarly, activation of dendritic cells in muscle-draining lymph nodes was strictly associated with the radio-sensitive cells expressing Tlr4. Altogether, these data suggest that MPL-mediated TLR4-signaling in hematopoietic cells is critical in the mode of action of AS01.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Células Madre Hematopoyéticas/inmunología , Lípido A/análogos & derivados , Saponinas/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/inmunología , Animales , Combinación de Medicamentos , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Lípido A/farmacología , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 4/genética , Vacunas/farmacología
4.
Infect Immun ; 85(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27872237

RESUMEN

Signaling through Toll-like receptors (TLRs), the main receptors in innate immunity, is essential for the defense of mucosal surfaces. It was previously shown that systemic TLR5 stimulation by bacterial flagellin induces an immediate, transient interleukin-22 (IL-22)-dependent antimicrobial response to bacterial or viral infections of the mucosa. This process was dependent on the activation of type 3 innate lymphoid cells (ILCs). The objective of the present study was to analyze the effects of flagellin treatment in a murine model of oral infection with Yersinia pseudotuberculosis (an invasive, Gram-negative, enteropathogenic bacterium that targets the small intestine). We found that systemic administration of flagellin significantly increased the survival rate after intestinal infection (but not systemic infection) by Y. pseudotuberculosis This protection was associated with a low bacterial count in the gut and the spleen. In contrast, no protection was afforded by administration of the TLR4 agonist lipopolysaccharide, suggesting the presence of a flagellin-specific effect. Lastly, we found that TLR5- and MyD88-mediated signaling was required for the protective effects of flagellin, whereas neither lymphoid cells nor IL-22 was involved.


Asunto(s)
Flagelina/inmunología , Interleucinas/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Flagelina/administración & dosificación , Interleucinas/genética , Mucosa Intestinal/microbiología , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , Proteínas Recombinantes de Fusión , Transducción de Señal , Receptores Toll-Like/metabolismo , Infecciones por Yersinia pseudotuberculosis/microbiología , Infecciones por Yersinia pseudotuberculosis/mortalidad , Interleucina-22
5.
J Infect Dis ; 210(3): 493-503, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24577508

RESUMEN

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


Asunto(s)
Interleucinas/metabolismo , Pulmón/metabolismo , Linfocitos/clasificación , Linfocitos/fisiología , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/metabolismo , Animales , Femenino , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucinas/genética , Activación de Linfocitos , Ratones , Ratones Noqueados , Streptococcus pneumoniae , Interleucina-22
6.
J Virol ; 87(12): 6911-24, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23596287

RESUMEN

Interleukin-22 (IL-22) has redundant, protective, or pathogenic functions during autoimmune, inflammatory, and infectious diseases. Here, we addressed the potential role of IL-22 in host defense and pathogenesis during lethal and sublethal respiratory H3N2 influenza A virus (IAV) infection. We show that IL-22, as well as factors associated with its production, are expressed in the lung tissue during the early phases of IAV infection. Our data indicate that retinoic acid receptor-related orphan receptor-γt (RORγt)-positive αß and γδ T cells, as well as innate lymphoid cells, expressed enhanced Il22 transcripts as early as 2 days postinfection. During lethal or sublethal IAV infections, endogenous IL-22 played no role in the control of IAV replication and in the development of the IAV-specific CD8(+) T cell response. During lethal infection, where wild-type (WT) mice succumbed to severe pneumonia, the lack of IL-22 did not accelerate or delay IAV-associated pathogenesis and animal death. In stark contrast, during sublethal IAV infection, IL-22-deficient animals had enhanced lung injuries and showed a lower airway epithelial integrity relative to WT littermates. Of importance, the protective effect of endogenous IL-22 in pulmonary damages was associated with a more controlled secondary bacterial infection. Indeed, after challenge with Streptococcus pneumoniae, IAV-experienced Il22(-/-) animals were more susceptible than WT controls in terms of survival rate and bacterial burden in the lungs. Together, IL-22 plays no major role during lethal influenza but is beneficial during sublethal H3N2 IAV infection, where it limits lung inflammation and subsequent bacterial superinfections.


Asunto(s)
Infecciones Bacterianas/inmunología , Coinfección/inmunología , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Interleucinas/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía/inmunología , Animales , Infecciones Bacterianas/microbiología , Coinfección/microbiología , Humanos , Subtipo H3N2 del Virus de la Influenza A/inmunología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/virología , Neumonía/patología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Streptococcus pneumoniae/patogenicidad , Interleucina-22
7.
Cell Host Microbe ; 32(3): 304-314.e8, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38417443

RESUMEN

Several vaccines targeting bacterial pathogens show reduced efficacy upon concurrent viral infection, indicating that a new vaccinology approach is required. To identify antigens for the human pathogen Streptococcus pneumoniae that are effective following influenza infection, we performed CRISPRi-seq in a murine model of superinfection and identified the conserved lafB gene as crucial for virulence. We show that LafB is a membrane-associated, intracellular protein that catalyzes the formation of galactosyl-glucosyl-diacylglycerol, a glycolipid important for cell wall homeostasis. Respiratory vaccination with recombinant LafB, in contrast to subcutaneous vaccination, was highly protective against S. pneumoniae serotypes 2, 15A, and 24F in a murine model. In contrast to standard capsule-based vaccines, protection did not require LafB-specific antibodies but was dependent on airway CD4+ T helper 17 cells. Healthy human individuals can elicit LafB-specific immune responses, indicating LafB antigenicity in humans. Collectively, these findings present a universal pneumococcal vaccine antigen that remains effective following influenza infection.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Infecciones Neumocócicas , Sobreinfección , Humanos , Animales , Ratones , Streptococcus pneumoniae , Infecciones Neumocócicas/prevención & control , Infecciones Neumocócicas/microbiología , Serogrupo , Células Th17 , Gripe Humana/prevención & control , Modelos Animales de Enfermedad , Vacunas Neumococicas , Antígenos Bacterianos/genética , Anticuerpos Antibacterianos
8.
EMBO Mol Med ; 16(1): 93-111, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177534

RESUMEN

Antimicrobial resistance is a global problem, rendering conventional treatments less effective and requiring innovative strategies to combat this growing threat. The tripartite AcrAB-TolC efflux pump is the dominant constitutive system by which Enterobacterales like Escherichia coli and Klebsiella pneumoniae extrude antibiotics. Here, we describe the medicinal chemistry development and drug-like properties of BDM91288, a pyridylpiperazine-based AcrB efflux pump inhibitor. In vitro evaluation of BDM91288 confirmed it to potentiate the activity of a panel of antibiotics against K. pneumoniae as well as revert clinically relevant antibiotic resistance mediated by acrAB-tolC overexpression. Using cryo-EM, BDM91288 binding to the transmembrane region of K. pneumoniae AcrB was confirmed, further validating the mechanism of action of this inhibitor. Finally, proof of concept studies demonstrated that oral administration of BDM91288 significantly potentiated the in vivo efficacy of levofloxacin treatment in a murine model of K. pneumoniae lung infection.


Asunto(s)
Antibacterianos , Proteínas de Escherichia coli , Animales , Ratones , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/farmacología , Klebsiella pneumoniae/metabolismo , Escherichia coli , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/farmacología
9.
J Infect Dis ; 206(5): 723-34, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22723642

RESUMEN

BACKGROUND: Exogenous activation of pulmonary invariant natural killer T (iNKT) cells, a population of lipid-reactive αß T lymphocytes, with use of mucosal α-galactosylceramide (α-GalCer) administration, is a promising approach to control respiratory bacterial infections. We undertook the present study to characterize mechanisms leading to α-GalCer-mediated protection against lethal infection with Streptococcus pneumoniae serotype 1, a major respiratory pathogen in humans. METHODS AND RESULTS: α-GalCer was administered by the intranasal route before infection with S. pneumoniae. We showed that respiratory dendritic cells (DCs), most likely the CD103(+) subset, play a major role in the activation (IFN-γ and IL-17 release) of pulmonary iNKT cells, whereas alveolar and interstitial macrophages are minor players. After challenge, S. pneumoniae was rapidly (4 hours) eliminated in the alveolar spaces, a phenomenon that depended on respiratory DCs and neutrophils, but not macrophages, and on the early production of both IFN-γ and IL-17. Protection was also associated with the synthesis of various interferon-dependent and IL-17-associated genes as revealed by transcriptomic analysis. CONCLUSIONS: These data imply a new function for pulmonary CD103(+) DCs in mucosal activation of iNKT cells and establish a critical role for both IFN-γ and IL-17 signalling pathways in mediating the innate immune response to S. pneumoniae.


Asunto(s)
Células Dendríticas/inmunología , Galactosilceramidas/farmacología , Células T Asesinas Naturales/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Animales , Antígenos CD/inmunología , Líquido del Lavado Bronquioalveolar/microbiología , Células Dendríticas/microbiología , Galactosilceramidas/uso terapéutico , Inmunidad Innata/inmunología , Cadenas alfa de Integrinas/inmunología , Interferón gamma/inmunología , Interleucina-17/inmunología , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos C57BL , Células T Asesinas Naturales/microbiología , Infecciones Neumocócicas/microbiología , Transducción de Señal
10.
PLoS Pathog ; 6(9): e1001097, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20844578

RESUMEN

Many enteropathogenic bacteria target the mammalian gut. The mechanisms protecting the host from infection are poorly understood. We have studied the protective functions of secretory antibodies (sIgA) and the microbiota, using a mouse model for S. typhimurium diarrhea. This pathogen is a common cause of diarrhea in humans world-wide. S. typhimurium (S. tm(att), sseD) causes a self-limiting gut infection in streptomycin-treated mice. After 40 days, all animals had overcome the disease, developed a sIgA response, and most had cleared the pathogen from the gut lumen. sIgA limited pathogen access to the mucosal surface and protected from gut inflammation in challenge infections. This protection was O-antigen specific, as demonstrated with pathogens lacking the S. typhimurium O-antigen (wbaP, S. enteritidis) and sIgA-deficient mice (TCRß(-/-)δ(-/-), J(H) (-/-), IgA(-/-), pIgR(-/-)). Surprisingly, sIgA-deficiency did not affect the kinetics of pathogen clearance from the gut lumen. Instead, this was mediated by the microbiota. This was confirmed using 'L-mice' which harbor a low complexity gut flora, lack colonization resistance and develop a normal sIgA response, but fail to clear S. tm(att) from the gut lumen. In these mice, pathogen clearance was achieved by transferring a normal complex microbiota. Thus, besides colonization resistance ( = pathogen blockage by an intact microbiota), the microbiota mediates a second, novel protective function, i.e. pathogen clearance. Here, the normal microbiota re-grows from a state of depletion and disturbed composition and gradually clears even very high pathogen loads from the gut lumen, a site inaccessible to most "classical" immune effector mechanisms. In conclusion, sIgA and microbiota serve complementary protective functions. The microbiota confers colonization resistance and mediates pathogen clearance in primary infections, while sIgA protects from disease if the host re-encounters the same pathogen. This has implications for curing S. typhimurium diarrhea and for preventing transmission.


Asunto(s)
Diarrea/prevención & control , Inmunoglobulina A Secretora/fisiología , Mucosa Intestinal/microbiología , Metagenoma/fisiología , Infecciones por Salmonella/prevención & control , Salmonella typhimurium/patogenicidad , Animales , Biomarcadores/metabolismo , Western Blotting , Diarrea/microbiología , Diarrea/patología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/fisiología , Humanos , Técnicas para Inmunoenzimas , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos O/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/patología , Salmonella typhimurium/genética , Salmonella typhimurium/inmunología , Estreptomicina/farmacología
11.
J Immunol ; 185(2): 1177-85, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20566828

RESUMEN

In adaptive immunity, Th17 lymphocytes produce the IL-17 and IL-22 cytokines that stimulate mucosal antimicrobial defenses and tissue repair. In this study, we observed that the TLR5 agonist flagellin induced swift and transient transcription of genes encoding IL-17 and IL-22 in lymphoid, gut, and lung tissues. This innate response also temporarily enhanced the expression of genes associated with the antimicrobial Th17 signature. The source of the Th17-related cytokines was identified as novel populations of CD3(neg)CD127(+) immune cells among which CD4-expressing cells resembling lymphoid tissue inducer cells. We also demonstrated that dendritic cells are essential for expression of Th17-related cytokines and so for stimulation of innate cells. These data define that TLR-induced activation of CD3(neg)CD127(+) cells and production of Th17-related cytokines may be crucial for the early defenses against pathogen invasion of host tissues.


Asunto(s)
Interleucina-17/inmunología , Interleucinas/inmunología , Membrana Mucosa/inmunología , Transducción de Señal/inmunología , Bazo/inmunología , Receptor Toll-Like 5/inmunología , Animales , Complejo CD3/genética , Complejo CD3/inmunología , Complejo CD3/metabolismo , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Flagelina/farmacología , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Íleon/efectos de los fármacos , Íleon/inmunología , Íleon/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Subunidad alfa del Receptor de Interleucina-7/genética , Subunidad alfa del Receptor de Interleucina-7/inmunología , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Membrana Mucosa/citología , Membrana Mucosa/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Bazo/citología , Bazo/metabolismo , Receptor Toll-Like 5/genética , Receptor Toll-Like 5/metabolismo , Interleucina-22
12.
Cell Host Microbe ; 29(1): 107-120.e6, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33120116

RESUMEN

Streptococcus pneumoniae is an opportunistic human pathogen that causes invasive diseases, including pneumonia, with greater health risks upon influenza A virus (IAV) co-infection. To facilitate pathogenesis studies in vivo, we developed an inducible CRISPR interference system that enables genome-wide fitness testing in one sequencing step (CRISPRi-seq). We applied CRISPRi-seq to assess bottlenecks and identify pneumococcal genes important in a murine pneumonia model. A critical bottleneck occurs at 48 h with few bacteria causing systemic infection. This bottleneck is not present during IAV superinfection, facilitating identification of pneumococcal pathogenesis-related genes. Top in vivo essential genes included purA, encoding adenylsuccinate synthetase, and the cps operon required for capsule production. Surprisingly, CRISPRi-seq indicated no fitness-related role for pneumolysin during superinfection. Interestingly, although metK (encoding S-adenosylmethionine synthetase) was essential in vitro, it was dispensable in vivo. This highlights advantages of CRISPRi-seq over transposon-based genetic screens, as all genes, including essential genes, can be tested for pathogenesis potential.


Asunto(s)
Genes Bacterianos , Neumonía Neumocócica/microbiología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Adenilosuccinato Sintasa/genética , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Femenino , Aptitud Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Virus de la Influenza A , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Operón , Infecciones por Orthomyxoviridae/complicaciones , Neumonía Neumocócica/complicaciones , Streptococcus pneumoniae/crecimiento & desarrollo , Sobreinfección
13.
Mucosal Immunol ; 14(1): 80-91, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32467605

RESUMEN

AU-rich element (ARE)-mediated mRNA decay represents a key mechanism to avoid excessive production of inflammatory cytokines. Tristetraprolin (TTP, encoded by Zfp36) is a major ARE-binding protein, since Zfp36-/- mice develop a complex multiorgan inflammatory syndrome that shares many features with spondyloarthritis. The role of TTP in intestinal homeostasis is not known. Herein, we show that Zfp36-/- mice do not develop any histological signs of gut pathology. However, they display a clear increase in intestinal inflammatory markers and discrete alterations in microbiota composition. Importantly, oral antibiotic treatment reduced both local and systemic joint and skin inflammation. We further show that absence of overt intestinal pathology is associated with local expansion of regulatory T cells. We demonstrate that this is related to increased vitamin A metabolism by gut dendritic cells, and identify RALDH2 as a direct target of TTP. In conclusion, these data bring insights into the interplay between microbiota-dependent gut and systemic inflammation during immune-mediated disorders, such as spondyloarthritis.


Asunto(s)
Aldehído Oxidorreductasas/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Homeostasis , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Tristetraprolina/metabolismo , Aldehído Oxidorreductasas/metabolismo , Animales , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Proteínas de Unión al ARN/metabolismo
14.
JCI Insight ; 6(5)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33497366

RESUMEN

Cancer is caused primarily by genomic alterations resulting in deregulation of gene regulatory circuits in key growth, apoptosis, or DNA repair pathways. Multiple genes associated with the initiation and development of tumors are also regulated at the level of mRNA decay, through the recruitment of RNA-binding proteins to AU-rich elements (AREs) located in their 3'-untranslated regions. One of these ARE-binding proteins, tristetraprolin (TTP; encoded by Zfp36), is consistently dysregulated in many human malignancies. Herein, using regulated overexpression or conditional ablation in the context of cutaneous chemical carcinogenesis, we show that TTP represents a critical regulator of skin tumorigenesis. We provide evidence that TTP controlled both tumor-associated inflammation and key oncogenic pathways in neoplastic epidermal cells. We identify Areg as a direct target of TTP in keratinocytes and show that EGFR signaling potentially contributed to exacerbated tumor formation. Finally, single-cell RNA-Seq analysis indicated that ZFP36 was downregulated in human malignant keratinocytes. We conclude that TTP expression by epidermal cells played a major role in the control of skin tumorigenesis.


Asunto(s)
Carcinogénesis/metabolismo , Queratinocitos/metabolismo , Neoplasias Cutáneas/metabolismo , Piel/metabolismo , Tristetraprolina/metabolismo , Regiones no Traducidas 3' , Elementos Ricos en Adenilato y Uridilato , Animales , Carcinogénesis/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Receptores ErbB/metabolismo , Redes Reguladoras de Genes , Humanos , Inflamación/metabolismo , Ratones Endogámicos C57BL , Estabilidad del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Neoplasias Cutáneas/genética
15.
Infect Immun ; 78(10): 4226-33, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20643849

RESUMEN

Streptococcus pneumoniae is a major cause of pneumonia in infants and the elderly. Innate defenses are essential to the control of pneumococcal infections, and deficient responses can trigger disease in susceptible individuals. Here we showed that flagellin can locally activate innate immunity and thereby increase the resistance to acute pneumonia. Flagellin mucosal treatment improved S. pneumoniae clearance in the lungs and promoted increased survival of infection. In addition, lung architecture was fully restored after the treatment of infected mice, indicating that flagellin allows the reestablishment of steady-state conditions. Using a flagellin mutant that is unable to signal through Toll-like receptor 5 (TLR5), we established that TLR5 signaling is essential for protection. In the respiratory tract, flagellin induced neutrophil infiltration into airways and upregulated the expression of genes coding for interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), CXCL1, CXCL2, and CCL20. Using depleting antibodies, we demonstrated that neutrophils are major effectors of protection. Further, we found that B- and T-cell-deficient SCID mice clear S. pneumoniae challenge to the same extent as immunocompetent animals, suggesting that these cell populations are not required for flagellin-induced protection. In conclusion, this study emphasizes that mucosal stimulation of innate immunity by a TLR not naturally engaged by S. pneumoniae can increase the potential to cure pneumococcal pneumonia.


Asunto(s)
Vacunas Bacterianas/inmunología , Flagelina/inmunología , Neumonía Neumocócica/prevención & control , Streptococcus pneumoniae/inmunología , Administración Intranasal , Animales , Linfocitos B , Vacunas Bacterianas/administración & dosificación , Lavado Broncoalveolar , Femenino , Flagelina/genética , Inmunidad Innata , Pulmón/citología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Mutación , Mucosa Nasal/inmunología , Neutrófilos/fisiología , Neumonía Neumocócica/inmunología , Transducción de Señal , Streptococcus pneumoniae/genética , Linfocitos T , Receptor Toll-Like 5/fisiología
16.
Commun Biol ; 2: 472, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31872076

RESUMEN

Monocytes play a major role in the defense against pathogens. They are rapidly mobilized to inflamed sites where they exert both proinflammatory and regulatory effector functions. It is still poorly understood how this dynamic and exceptionally plastic system is controlled at the molecular level. Herein, we evaluated the differentiation process that occurs in Ly6Chi monocytes during oral infection by Toxoplasma gondii. Flow cytometry and single-cell analysis revealed distinct activation status and gene expression profiles in the bone marrow, the spleen and the lamina propria of infected mice. We provide further evidence that acquisition of effector functions, such as the capacity to produce interleukin-27, is accompanied by distinct waves of epigenetic programming, highlighting a role for STAT1/IRF1 in the bone marrow and AP-1/NF-κB in the periphery. This work broadens our understanding of the molecular events that occur in vivo during monocyte differentiation in response to inflammatory cues.


Asunto(s)
Diferenciación Celular/inmunología , Monocitos/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Toxoplasmosis/parasitología , Animales , Reprogramación Celular/genética , Biología Computacional/métodos , Epigénesis Genética , Perfilación de la Expresión Génica , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Monocitos/citología , Monocitos/metabolismo , Análisis de la Célula Individual , Toxoplasmosis/genética , Toxoplasmosis/metabolismo
17.
Front Immunol ; 9: 1856, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30147700

RESUMEN

Allergic asthma is a chronic Th2 inflammatory disease of the lower airways affecting a growing number of people worldwide. The impact of infections and microbiota composition on allergic asthma has been investigated frequently. Until now, however, there have been few attempts to investigate the impact of asthma on the control of infectious microorganisms and the underlying mechanisms. In this work, we characterize the consequences of allergic asthma on intranasal (i.n.) infection by Brucella bacteria in mice. We observed that i.n. sensitization with extracts of the house dust mite Dermatophagoides farinae or the mold Alternaria alternata (Alt) significantly increased the number of Brucella melitensis, Brucella suis, and Brucella abortus in the lungs of infected mice. Microscopic analysis showed dense aggregates of infected cells composed mainly of alveolar macrophages (CD11c+ F4/80+ MHCII+) surrounded by neutrophils (Ly-6G+). Asthma-induced Brucella susceptibility appears to be dependent on CD4+ T cells, the IL-4/STAT6 signaling pathway and IL-10, and is maintained in IL-12- and IFN-γR-deficient mice. The effects of the Alt sensitization protocol were also tested on Streptococcus pneumoniae and Mycobacterium tuberculosis pulmonary infections. Surprisingly, we observed that Alt sensitization strongly increases the survival of S. pneumoniae infected mice by a T cell and STAT6 independent signaling pathway. In contrast, the course of M. tuberculosis infection is not affected in the lungs of sensitized mice. Our work demonstrates that the impact of the same allergic sensitization protocol can be neutral, negative, or positive with regard to the resistance of mice to bacterial infection, depending on the bacterial species.


Asunto(s)
Asma/inmunología , Brucella/fisiología , Brucelosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Hipersensibilidad/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Alternaria/inmunología , Animales , Antígenos Dermatofagoides/inmunología , Antígenos Fúngicos/inmunología , Asma/microbiología , Dermatophagoides farinae/inmunología , Hipersensibilidad/microbiología , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Pulmón/microbiología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal
18.
JCI Insight ; 2(11)2017 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-28570274

RESUMEN

Tristetraprolin (TTP, encoded by the Zfp36 gene) regulates the mRNA stability of several important cytokines. Due to the critical role of this RNA-binding protein in the control of inflammation, TTP deficiency leads to the spontaneous development of a complex inflammatory syndrome. So far, this phenotype has been largely attributed to dysregulated production of TNF and IL­23 by myeloid cells, such as macrophages or DCs. Here, we generated mice with conditional deletion of TTP in keratinocytes (Zfp36fl/flK14-Cre mice, referred to herein as Zfp36ΔEP mice). Unlike DC-restricted (CD11c-Cre) or myeloid cell-restricted (LysM-Cre) TTP ablation, these mice developed exacerbated inflammation in the imiquimod-induced psoriasis model. Furthermore, Zfp36ΔEP mice progressively developed a spontaneous pathology with systemic inflammation, psoriatic-like skin lesions, and dactylitis. Finally, we provide evidence that keratinocyte-derived TNF production drives these different pathological features. In summary, these findings expand current views on the initiation of psoriasis and related arthritis by revealing the keratinocyte-intrinsic role of TTP.

19.
Sci Rep ; 6: 39475, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27996000

RESUMEN

Saponins represent a promising class of vaccine adjuvant. Together with the TLR4-ligand MPL, QS-21 is part of the Adjuvant System AS01, a key component of the malaria and zoster candidate vaccines that display demonstrated clinical efficacy. However, the mechanism of action of QS-21 in this liposomal formulation is poorly understood. Upon intra-muscular immunisation, we observed that QS-21 rapidly accumulated in CD169+ resident macrophages of the draining lymph node where it elicited a local innate immune response. Depletion of these cells abrogated QS-21-mediated innate cell recruitment to the lymph node, dendritic cell (DC) phenotypic maturation as well as the adjuvant effect on T-cell and antibody responses to co-administered antigens. DCs rather than lymph node-resident macrophages were directly involved in T-cell priming by QS-21, as revealed by the decrease in antigen-specific T-cell response in Batf3-/- mice. Further analysis showed that the adjuvant effect of QS-21 depended on the integration of Caspase-1 and MyD88 pathways, at least in part through the local release of HMGB1. Taken together, this work unravels the key role of lymph node sentinel macrophage in controlling the adjuvant effect of a molecule proven to improve vaccine response in humans.

20.
Vaccine ; 33(29): 3331-41, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-26003491

RESUMEN

The Toll-like receptor 5 (TLR5) agonist flagellin is an effective adjuvant for vaccination. Recently, we demonstrated that the adaptive responses stimulated by intranasal administration of flagellin and antigen were linked to TLR5 signaling in the lung epithelium. The present study sought to identify the antigen presenting cells involved in this adjuvant activity. We first found that the lung dendritic cells captured antigen very efficiently in a process independent of TLR5. However, TLR5-mediated signaling specifically enhanced the maturation of lung dendritic cells. Afterward, the number of antigen-bound and activated conventional dendritic cells (both CD11b(+) and CD103(+)) increased in the mediastinal lymph nodes in contrast to monocyte-derived dendritic cells. These data suggested that flagellin-activated lung conventional dendritic cells migrate to the draining lymph nodes. The lymph node dendritic cells, in particular CD11b(+) cells, were essential for induction of CD4 T-cell response. Lastly, neutrophils and monocytes were recruited into the lungs by flagellin administration but did not contribute to the adjuvant activity. The functional activation of conventional dendritic cells was independent of direct TLR5 signaling, thereby supporting the contribution of maturation signals produced by flagellin-stimulated airway epithelium. In conclusion, our results demonstrated that indirect TLR5-dependent stimulation of airway conventional dendritic cells is essential to flagellin's mucosal adjuvant activity.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Flagelina/metabolismo , Mucosa Respiratoria/inmunología , Sistema Respiratorio/inmunología , Receptor Toll-Like 5/agonistas , Animales , Inmunidad Innata , Inmunidad Mucosa , Ratones Endogámicos BALB C , Ratones Endogámicos C3H
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda