Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(8): e1011575, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37603560

RESUMEN

Mycobacterium abscessus causes severe disease in patients with cystic fibrosis. Little is known in M. abscessus about the roles of small regulatory RNAs (sRNA) in gene regulation. We show that the sRNA B11 controls gene expression and virulence-associated phenotypes in this pathogen. B11 deletion from the smooth strain ATCC_19977 produced a rough strain, increased pro-inflammatory signaling and virulence in multiple infection models, and increased resistance to antibiotics. Examination of clinical isolate cohorts identified isolates with B11 mutations or reduced expression. We used RNAseq and proteomics to investigate the effects of B11 on gene expression and test the impact of mutations found in clinical isolates. Over 200 genes were differentially expressed in the deletion mutant. Strains with the clinical B11 mutations showed expression trends similar to the deletion mutant, suggesting partial loss of function. Among genes upregulated in the B11 mutant, there was a strong enrichment for genes with B11-complementary sequences in their predicted ribosome binding sites (RBS), consistent with B11 functioning as a negative regulator that represses translation via base-pairing to RBSs. Comparing the proteomes similarly revealed that upregulated proteins were strongly enriched for B11-complementary sequences. Intriguingly, genes upregulated in the absence of B11 included components of the ESX-4 secretion system, critical for M. abscessus virulence. Many of these genes had B11-complementary sequences at their RBSs, which we show is sufficient to mediate repression by B11 through direct binding. Altogether, our data show that B11 acts as a direct negative regulator and mediates (likely indirect) positive regulation with pleiotropic effects on gene expression and clinically important phenotypes in M. abscessus. The presence of hypomorphic B11 mutations in clinical strains is consistent with the idea that lower B11 activity may be advantageous for M. abscessus in some clinical contexts. This is the first report on an sRNA role in M. abscessus.


Asunto(s)
Mycobacterium abscessus , ARN Pequeño no Traducido , Mycobacterium abscessus/genética , Virulencia/genética , Antibacterianos , ARN Pequeño no Traducido/genética
2.
Int J Mol Sci ; 21(18)2020 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-32916885

RESUMEN

Pulmonary infections caused by Mycobacterium abscessus (MA) have increased over recent decades, affecting individuals with underlying pathologies such as chronic obstructive pulmonary disease, bronchiectasis and, especially, cystic fibrosis. The lack of a representative and standardized model of chronic infection in mice has limited steps forward in the field of MA pulmonary infection. To overcome this challenge, we refined the method of agar beads to establish MA chronic infection in immunocompetent mice. We evaluated bacterial count, lung pathology and markers of inflammation and we performed longitudinal studies with magnetic resonance imaging (MRI) up to three months after MA infection. In this model, MA was able to establish a persistent lung infection for up to two months and with minimal systemic spread. Lung histopathological analysis revealed granulomatous inflammation around bronchi characterized by the presence of lymphocytes, aggregates of vacuolated histiocytes and a few neutrophils, mimicking the damage observed in humans. Furthermore, MA lung lesions were successfully monitored for the first time by MRI. The availability of this murine model and the introduction of the successfully longitudinal monitoring of the murine lung lesions with MRI pave the way for further investigations on the impact of MA pathogenesis and the efficacy of novel treatments.


Asunto(s)
Modelos Animales de Enfermedad , Pulmón/patología , Infecciones por Mycobacterium no Tuberculosas/patología , Mycobacterium abscessus , Neumonía Bacteriana/patología , Animales , Enfermedad Crónica , Pulmón/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Ratones Endogámicos C57BL , Infecciones por Mycobacterium no Tuberculosas/diagnóstico por imagen , Neumonía Bacteriana/diagnóstico por imagen
3.
Mamm Genome ; 29(7-8): 550-557, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29947963

RESUMEN

Understanding the significance of human genetic diversity in modulating host susceptibility to opportunistic infections is an emerging challenge in the field of respiratory illnesses. While it is recognized that diverse bacterial strains account for differential disease manifestations, emerging data indicate that host genetic diversity is an important determinant factor that influences the severity of opportunistic infections. With particular regard to respiratory illnesses mediated by the gram-negative bacterium Pseudomonas aeruginosa, diverse genetic background is also emerging as a key contributor. Human-genome-wide association studies are a common approach for determining the inter-individual genetic variation associated with variability of the pulmonary infections. Historically, diverse murine inbred mouse strains and ex-vivo cellular models were considered complementary to human studies for establishing the contribution of genetic background to P. aeruginosa respiratory infections. More recently, the development of a new mouse model of infection, mirroring human airway diseases, combined with innovative murine resource populations, modelling human genetic variation, provides additional insights into the mechanisms of genetic susceptibility. In this review, we cover the recent state of the art of human and animal studies and we discuss future potential challenges in the field of P. aeruginosa respiratory infections.


Asunto(s)
Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno/genética , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/fisiología , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/microbiología , Animales , Modelos Animales de Enfermedad , Antecedentes Genéticos , Heterogeneidad Genética , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Ratones Endogámicos , Sitios de Carácter Cuantitativo
4.
Int J Mol Sci ; 19(1)2018 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-29315274

RESUMEN

Several chronic respiratory diseases are characterized by recurrent and/or persistent infections, chronic inflammatory responses and tissue remodeling, including increased levels of glycosaminoglycans which are known structural components of the airways. Among glycosaminoglycans, heparan sulfate (HS) has been suggested to contribute to excessive inflammatory responses. Here, we aim at (i) investigating whether long-term infection by Pseudomonas aeruginosa, one of the most worrisome threat in chronic respiratory diseases, may impact HS levels, and (ii) exploring HS competitors as potential anti-inflammatory drugs during P. aeruginosa pneumonia. P. aeruginosa clinical strains and ad-hoc synthesized HS competitors were used in vitro and in murine models of lung infection. During long-term chronic P. aeruginosa colonization, infected mice showed higher heparin/HS levels, evaluated by high performance liquid chromatography-mass spectrometry after selective enzymatic digestion, compared to uninfected mice. Among HS competitors, an N-acetyl heparin and a glycol-split heparin dampened leukocyte recruitment and cytokine/chemokine production induced by acute and chronic P. aeruginosa pneumonia in mice. Furthermore, treatment with HS competitors reduced bacterial burden during chronic murine lung infection. In vitro, P. aeruginosa biofilm formation decreased upon treatment with HS competitors. Overall, these findings support further evaluation of HS competitors as a novel therapy to counteract inflammation and infection during P. aeruginosa pneumonia.


Asunto(s)
Antiinflamatorios/uso terapéutico , Heparitina Sulfato/química , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/fisiología , Infecciones del Sistema Respiratorio/prevención & control , Animales , Antiinflamatorios/síntesis química , Antiinflamatorios/farmacología , Biopelículas/efectos de los fármacos , Quimiocinas/análisis , Quimiocinas/metabolismo , Cromatografía Líquida de Alta Presión , Citocinas/análisis , Citocinas/metabolismo , Modelos Animales de Enfermedad , Heparitina Sulfato/análisis , Heparitina Sulfato/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/aislamiento & purificación , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/microbiología
6.
J Biol Chem ; 290(35): 21305-19, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26160169

RESUMEN

Lung infection by Burkholderia species, in particular Burkholderia cenocepacia, accelerates tissue damage and increases post-lung transplant mortality in cystic fibrosis patients. Host-microbe interplay largely depends on interactions between pathogen-specific molecules and innate immune receptors such as Toll-like receptor 4 (TLR4), which recognizes the lipid A moiety of the bacterial lipopolysaccharide (LPS). The human TLR4·myeloid differentiation factor 2 (MD-2) LPS receptor complex is strongly activated by hexa-acylated lipid A and poorly activated by underacylated lipid A. Here, we report that B. cenocepacia LPS strongly activates human TLR4·MD-2 despite its lipid A having only five acyl chains. Furthermore, we show that aminoarabinose residues in lipid A contribute to TLR4-lipid A interactions, and experiments in a mouse model of LPS-induced endotoxic shock confirmed the proinflammatory potential of B. cenocepacia penta-acylated lipid A. Molecular modeling combined with mutagenesis of TLR4-MD-2 interactive surfaces suggests that longer acyl chains and the aminoarabinose residues in the B. cenocepacia lipid A allow exposure of the fifth acyl chain on the surface of MD-2 enabling interactions with TLR4 and its dimerization. Our results provide a molecular model for activation of the human TLR4·MD-2 complex by penta-acylated lipid A explaining the ability of hypoacylated B. cenocepacia LPS to promote proinflammatory responses associated with the severe pathogenicity of this opportunistic bacterium.


Asunto(s)
Infecciones por Burkholderia/inmunología , Burkholderia cenocepacia/inmunología , Lípido A/inmunología , Antígeno 96 de los Linfocitos/inmunología , Receptor Toll-Like 4/inmunología , Acilación , Animales , Burkholderia cenocepacia/química , Burkholderia cenocepacia/aislamiento & purificación , Línea Celular , Células HEK293 , Humanos , Inmunidad Innata , Inflamación/inmunología , Inflamación/microbiología , Interleucina-6/inmunología , Lípido A/química , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular
7.
BMC Genomics ; 17: 351, 2016 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-27169516

RESUMEN

BACKGROUND: P. aeruginosa is one of the top three causes of opportunistic human bacterial infections. The remarkable variability in the clinical outcomes of this infection is thought to be associated with genetic predisposition. However, the genes underlying host susceptibility to P. aeruginosa infection are still largely unknown. RESULTS: As a step towards mapping these genes, we applied a genome wide linkage analysis approach to a mouse model. A large F2 intercross population, obtained by mating P. aeruginosa-resistant C3H/HeOuJ, and susceptible A/J mice, was used for quantitative trait locus (QTL) mapping. The F2 progenies were challenged with a P. aeruginosa clinical strain and monitored for the survival time up to 7 days post-infection, as a disease phenotype associated trait. Selected phenotypic extremes of the F2 distribution were genotyped with high-density single nucleotide polymorphic (SNP) markers, and subsequently QTL analysis was performed. A significant locus was mapped on chromosome 6 and was named P . aeruginosa infection resistance locus 1 (Pairl1). The most promising candidate genes, including Dok1, Tacr1, Cd207, Clec4f, Gp9, Gata2, Foxp1, are related to pathogen sensing, neutrophils and macrophages recruitment and inflammatory processes. CONCLUSIONS: We propose a set of genes involved in the pathogenesis of P. aeruginosa infection that may be explored to complement human studies.


Asunto(s)
Mapeo Cromosómico/métodos , Redes Reguladoras de Genes , Infecciones por Pseudomonas/genética , Pseudomonas aeruginosa/fisiología , Animales , Modelos Animales de Enfermedad , Ligamiento Genético , Predisposición Genética a la Enfermedad , Humanos , Ratones , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo
9.
Antimicrob Agents Chemother ; 59(12): 7265-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26369968

RESUMEN

Trimethoprim-sulfamethoxazole (SXT) is a possible alternative for the treatment of community- and hospital-acquired methicillin-resistant Staphylococcus aureus (MRSA) due to the susceptibility of most MRSA strains to the drug. However, after long-term treatment with SXT, thymidine-dependent (TD) SXT-resistant small-colony variants (SCVs) emerge. In TD-SCVs, mutations of thymidylate synthase ([TS] thyA) occur. Until now, it has never been systematically investigated that SXT is triggering the induction and/or selection of TD-SCVs. In our study, we performed induction, reversion, and competition experiments in vitro and in vivo using a chronic mouse pneumonia model to determine the impact of SXT on the emergence of TD-SCVs. SCVs were characterized by light and transmission electron microscopy (TEM) and auxotrophism testing. Short-term exposure of S. aureus to SXT induced the TD-SCV phenotype in S. aureus SH1000, while selection of TD-SCVs with thyA mutations occurred after long-term exposure. In reversion experiments with clinical and laboratory TD-SCVs, all revertants carried compensating mutations at the initially identified mutation site. Competition experiments in vitro and in vivo revealed a survival and growth advantage of the ΔthyA mutant under low-thymidine availability and SXT exposure although this advantage was less profound in vivo. Our results show that SXT induces the TD-SCV phenotype after short-term exposure, while long-term exposure selects for thyA mutations, which provide an advantage for TD-SCVs under specified conditions. Thus, our results further an understanding of the dynamic processes occurring during SXT exposure with induction and selection of S. aureus TD-SCVs.


Asunto(s)
Antibacterianos/efectos adversos , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Timidilato Sintasa/genética , Combinación Trimetoprim y Sulfametoxazol/efectos adversos , Animales , Proteínas Bacterianas/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana/genética , Expresión Génica , Aptitud Genética/efectos de los fármacos , Masculino , Staphylococcus aureus Resistente a Meticilina/enzimología , Staphylococcus aureus Resistente a Meticilina/genética , Ratones , Ratones Endogámicos C57BL , Mutación , Neumonía Bacteriana/tratamiento farmacológico , Selección Genética/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Timidina/metabolismo , Timidilato Sintasa/deficiencia
10.
BMC Genet ; 16: 106, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26310945

RESUMEN

BACKGROUND: Pseudomonas aeruginosa is one of the top three causes of opportunistic infections in humans. Patients with a compromised immune system, due to immunosuppressive therapies or underlying diseases such as cancer, AIDS or the hereditary disease cystic fibrosis, are at risk of developing P. aeruginosa infection. However, clinical evidence indicates extremely variable outcomes of P. aeruginosa infections in individuals at risk, suggesting that host multi-complex genetic traits may influence the severity of this opportunistic infection. Here, we have used an innovative experimental model to dissect whether host genetic background, such as those found in the outbred population, could influence the risk of morbidity and mortality to P. aeruginosa pneumonia. RESULTS: A highly genetically-diverse mouse resource population, Collaborative Cross (CC) mice, was infected with a clinical strain of P. aeruginosa and subsequently monitored for mortality, mean survival time, and morbidity, change in body weight for seven days post infection. Disease phenotypes ranged from complete resistance and recovery of body weight to lethal disease. Initial variables, including body weight, age and gender, have limited influence on P. aeruginosa outcome, emphasizing the role of host genetic background in defining the risk of morbidity and mortality. When broad-sense heritability of phenotypic traits was evaluated, it confirmed the influence of genetic profile rather than environmental factors among the CC lines during P. aeruginosa infection. CONCLUSION: This innovative model system can potentially reproduce the variables responses of disease severity observed in humans during P. aeruginosa pneumonia. Our results demonstrated that a widely-marked differential response to P. aeruginosa airway infection in term of morbidity and mortality, is mainly affected by host genetic factors, as multiple genetic loci or polymorphic variations.


Asunto(s)
Variación Genética , Interacciones Huésped-Patógeno/genética , Neumonía Bacteriana/genética , Neumonía Bacteriana/microbiología , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa , Factores de Edad , Animales , Peso Corporal , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Masculino , Ratones , Fenotipo , Neumonía Bacteriana/diagnóstico , Infecciones por Pseudomonas/diagnóstico , Carácter Cuantitativo Heredable , Índice de Severidad de la Enfermedad , Factores Sexuales
11.
Cells ; 11(7)2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35406809

RESUMEN

Cystic Fibrosis (CF) is an autosomal recessive disease caused by mutations in the gene encoding for the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) protein, expressed on the apical surface of epithelial cells. CFTR absence/dysfunction results in ion imbalance and airway surface dehydration that severely compromise the CF airway microenvironment, increasing infection susceptibility. Recently, novel therapies aimed at correcting the basic CFTR defect have become available, leading to substantial clinical improvement of CF patients. The restoration or increase of CFTR function affects the airway microenvironment, improving local defence mechanisms. CFTR modulator drugs might therefore affect the development of chronic airway infections and/or improve the status of existing infections in CF. Thus far, however, the full extent of these effects of CFTR-modulators, especially in the long-term remains still unknown. This review aims to provide an overview of current evidence on the potential impact of CFTR modulators on airway infections in CF. Their role in affecting CF microbiology, the susceptibility to infections as well as the potential efficacy of their use in preventing/decreasing the development of chronic lung infections and the recurrent acute exacerbations in CF will be critically analysed.


Asunto(s)
Fibrosis Quística , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Transporte Iónico , Mutación/genética , Sistema Respiratorio/metabolismo
12.
J Biomed Biotechnol ; 2011: 852513, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21785567

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen and causes a wide range of acute and chronic infections. P. aeruginosa infections are kept in check by an effective immune surveillance in the healthy host, while any imbalance or defect in the normal immune response can manifest in disease. Invasive acute infection in the immunocompromised patients is mediated by potent extracellular and cell bound bacterial virulence factors. Life-threatening chronic infection in cystic fibrosis patients is maintained by pathogenic variants that contribute to evade detection and clearance by the immune system. Here, we reviewed the molecular basis of receptor-mediated recognition of P. aeruginosa and their role in initiating inflammation and the colonization. In addition, the consequence of the P. aeruginosa genetic adaptation for the antibacterial defence and the maintaining of chronic infection are discussed.

13.
mBio ; 11(2)2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127447

RESUMEN

Human genetics influence a range of pathological and clinical phenotypes in respiratory infections; however, the contributions of disease modifiers remain underappreciated. We exploited the Collaborative Cross (CC) mouse genetic-reference population to map genetic modifiers that affect the severity of Pseudomonas aeruginosa lung infection. Screening for P. aeruginosa respiratory infection in a cohort of 39 CC lines exhibits distinct disease phenotypes ranging from complete resistance to lethal disease. Based on major changes in the survival times, a quantitative-trait locus (QTL) was mapped on murine chromosome 3 to the genomic interval of Mb 110.4 to 120.5. Within this locus, composed of 31 protein-coding genes, two candidate genes, namely, dihydropyrimidine dehydrogenase (Dpyd) and sphingosine-1-phosphate receptor 1 (S1pr1), were identified according to the level of genome-wide significance and disease gene prioritization. Functional validation of the S1pr1 gene by pharmacological targeting in C57BL/6NCrl mice confirmed its relevance in P. aeruginosa pathophysiology. However, in a cohort of Canadian patients with cystic fibrosis (CF) disease, regional genetic-association analysis of the syntenic human locus on chromosome 1 (Mb 97.0 to 105.0) identified two single-nucleotide polymorphisms (rs10875080 and rs11582736) annotated to the Dpyd gene that were significantly associated with age at first P. aeruginosa infection. Thus, there is evidence that both genes might be implicated in this disease. Our results demonstrate that the discovery of murine modifier loci may generate information that is relevant to human disease progression.IMPORTANCE Respiratory infection caused by P. aeruginosa is one of the most critical health burdens worldwide. People affected by P. aeruginosa infection include patients with a weakened immune system, such as those with cystic fibrosis (CF) genetic disease or non-CF bronchiectasis. Disease outcomes range from fatal pneumonia to chronic life-threatening infection and inflammation leading to the progressive deterioration of pulmonary function. The development of these respiratory infections is mediated by multiple causes. However, the genetic factors underlying infection susceptibility are poorly known and difficult to predict. Our study employed novel approaches and improved mouse disease models to identify genetic modifiers that affect the severity of P. aeruginosa lung infection. We identified candidate genes to enhance our understanding of P. aeruginosa infection in humans and provide a proof of concept that could be exploited for other human pathologies mediated by bacterial infection.


Asunto(s)
Ratones de Colaboración Cruzada/genética , Predisposición Genética a la Enfermedad , Pulmón/microbiología , Infecciones por Pseudomonas/genética , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/microbiología , Adolescente , Animales , Línea Celular Tumoral , Niño , Cromosomas , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Adulto Joven
15.
J Cyst Fibros ; 17(2S): S40-S45, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29107600

RESUMEN

Cystic Fibrosis (CF) lung disease is associated with dysregulation of host defence systems, which ultimately disrupts the balance between inflammation and resolution and leaves the host susceptible to repeated infection. However, the mechanisms underlying these defects are complex and continue to garner significant interest among the CF research community. This review explores emerging data on novel aspects of innate host defence with promising biomarker and therapeutic potential for CF lung disease. Improved understanding of inflammation and host defence against pathogens in patients and animal models during the progression of CF lung disease is pivotal for the discovery of new therapeutics that can limit and/or prevent damage from birth.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Fibrosis Quística/inmunología , Fibrosis Quística/metabolismo , Humanos , Infecciones/inmunología , Inflamación
16.
Methods Mol Biol ; 1488: 551-578, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27933544

RESUMEN

Infection is one of the leading causes of human mortality and morbidity. Exposure to microbial agents is obviously required. However, also non-microbial environmental and host factors play a key role in the onset, development and outcome of infectious disease, resulting in large of clinical variability between individuals in a population infected with the same microbe. Controlled and standardized investigations of the genetics of susceptibility to infectious disease are almost impossible to perform in humans whereas mouse models allow application of powerful genomic techniques to identify and validate causative genes underlying human diseases with complex etiologies. Most of current animal models used in complex traits diseases genetic mapping have limited genetic diversity. This limitation impedes the ability to create incorporated network using genetic interactions, epigenetics, environmental factors, microbiota, and other phenotypes. A novel mouse genetic reference population for high-resolution mapping and subsequently identifying genes underlying the QTL, namely the Collaborative Cross (CC) mouse genetic reference population (GRP) was recently developed. In this chapter, we discuss a variety of approaches using CC mice for mapping genes underlying quantitative trait loci (QTL) to dissect the host response to polygenic traits, including infectious disease caused by bacterial agents and its toxins.


Asunto(s)
Infecciones Bacterianas/etiología , Infecciones Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno/genética , Animales , Infecciones Bacterianas/mortalidad , Mapeo Cromosómico , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Variación Genética , Ratones , Ratones Endogámicos , Fenotipo , Sitios de Carácter Cuantitativo
17.
Cytokine Growth Factor Rev ; 30: 19-27, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27033174

RESUMEN

The interleukin (IL)-17A/IL-17 receptor A (IL-17RA) axis is emerging as a key player in host defence. Several studies have demonstrated that IL-17A-mediated responses play a critical role in both acute and chronic inflammation induced by infectious agents, environmental stimuli and genetic diseases in the airways. In this regard, it is becoming evident that IL-17A/IL-17RA signalling may have a protective and beneficial impact on health, but that it can also result in detrimental outcomes. On one hand, the IL-17A/IL-17RA axis can contribute to the elimination of noxious stimuli and to the resolution of acute inflammatory processes; on the other hand, it can exacerbate immunopathological responses, contributing to the development and progression of chronic respiratory illnesses. In addition, cellular and molecular signatures underlying IL-17A/IL-17RA signalling have been increasingly identified, although further studies are needed to clarify such complex responses. Here, we discuss the latest discoveries on the role of the IL-17A/IL-17RA axis in driving host pulmonary defence and immunopathology.


Asunto(s)
Interleucina-17/inmunología , Pulmón/inmunología , Receptores de Interleucina-17/inmunología , Animales , Humanos , Enfermedades Pulmonares/inmunología , Infecciones del Sistema Respiratorio/inmunología , Transducción de Señal
18.
Sci Rep ; 6: 21465, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26883959

RESUMEN

Repeated cycles of infections, caused mainly by Pseudomonas aeruginosa, combined with a robust host immune response and tissue injury, determine the course and outcome of cystic fibrosis (CF) lung disease. As the disease progresses, P. aeruginosa adapts to the host modifying dramatically its phenotype; however, it remains unclear whether and how bacterial adaptive variants and their persistence influence the pathogenesis and disease development. Using in vitro and murine models of infection, we showed that P. aeruginosa CF-adaptive variants shaped the innate immune response favoring their persistence. Next, we refined a murine model of chronic pneumonia extending P. aeruginosa infection up to three months. In this model, including CFTR-deficient mice, we unveil that the P. aeruginosa persistence lead to CF hallmarks of airway remodelling and fibrosis, including epithelial hyperplasia and structure degeneration, goblet cell metaplasia, collagen deposition, elastin degradation and several additional markers of tissue damage. This murine model of P. aeruginosa chronic infection, reproducing CF lung pathology, will be instrumental to identify novel molecular targets and test newly tailored molecules inhibiting chronic inflammation and tissue damage processes in pre-clinical studies.


Asunto(s)
Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Animales , Línea Celular , Quimiocinas/metabolismo , Fibrosis Quística/complicaciones , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Infecciones Oportunistas , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/patología
19.
Sci Rep ; 6: 36924, 2016 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-27848994

RESUMEN

Patients with P. aeruginosa airways infection show markedly variable clinical phenotypes likely influenced by genetic backgrounds. Here, we investigated the cellular events involved in resistance and susceptibility to P. aeruginosa chronic infection using genetically distinct inbred mouse strains. As for patients, different murine genotypes revealed variable susceptibility to infection. When directly compared, resistant C3H/HeOuJ and susceptible A/J strains revealed distinct immune responsiveness to the pathogen. In C3H/HeOuJ resistant mice, IL17-producing cells rapidly and transiently infiltrated the infected lung, and this was paralleled by the acute accumulation of alveolar macrophages, bacterial clearance and resolution of infection. In contrast, A/J susceptible mice revealed a more delayed and prolonged lung infiltration by IL17+ and IFNγ+ cells, persistence of innate inflammatory cells and establishment of chronic infection. We conclude that the host genetic background confers diverse immunoreactivity to P. aeruginosa and IL17-producing cells might contribute to the progress of chronic lung infection.


Asunto(s)
Resistencia a la Enfermedad , Predisposición Genética a la Enfermedad , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/inmunología , Animales , Antecedentes Genéticos , Interleucina-17/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Pulmón/patología , Ratones
20.
Sci Rep ; 6: 25937, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27189736

RESUMEN

Resistance and tolerance mechanisms participate to the interplay between host and pathogens. IL-17-mediated response has been shown to be crucial for host resistance to respiratory infections, whereas its role in host tolerance during chronic airway colonization is still unclear. Here, we investigated whether IL-17-mediated response modulates mechanisms of host tolerance during airways chronic infection by P. aeruginosa. First, we found that IL-17A levels were sustained in mice at both early and advanced stages of P. aeruginosa chronic infection and confirmed these observations in human respiratory samples from cystic fibrosis patients infected by P. aeruginosa. Using IL-17a(-/-) or IL-17ra(-/-) mice, we found that the deficiency of IL-17A/IL-17RA axis was associated with: i) increased incidence of chronic infection and bacterial burden, indicating its role in the host resistance to P. aeruginosa; ii) reduced cytokine levels (KC), tissue innate immune cells and markers of tissue damage (pro-MMP-9, elastin degradation, TGF-ß1), proving alteration of host tolerance. Blockade of IL-17A activity by a monoclonal antibody, started when chronic infection is established, did not alter host resistance but increased tolerance. In conclusion, this study identifies IL-17-mediated response as a negative regulator of host tolerance during P. aeruginosa chronic airway infection.


Asunto(s)
Fibrosis Quística/microbiología , Interleucina-17/genética , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/patogenicidad , Infecciones del Sistema Respiratorio/microbiología , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata , Incidencia , Interleucina-17/metabolismo , Ratones , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/inmunología , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/inmunología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA