Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Lung ; 202(3): 281-289, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38713421

RESUMEN

INTRODUCTION: Immunoglobulins play a vital role in host immune response and in the pathogenesis of conditions like asthma. Therapeutic agents such as monoclonal antibodies target specific elements of the asthmatic inflammatory cascade. Decisions to utilize these medications are often based on systemic inflammatory profiling without direct insight into the airway inflammatory profile. We sought to investigate the relationship between immunoglobulin and cytokine profiles in the airway and systemic immune compartments of adult asthmatics. METHODS: Blood sampling and bronchoscopy with bronchoalveolar lavage (BAL) were performed in 76 well-defined adult asthmatics. Antibody and cytokine profiles were measured in both BAL and serum using ELISA and quantibody arrays. RESULTS: There was no relationship between BAL and serum levels of IgE. This is of significance in an asthma population. For some analytes, correlation analysis was significant (P < 0.05) indicating representativeness of our cohort and experimental setup in those cases. Nevertheless, the predictive power (r2) of the BAL-to-serum comparisons was mostly low except for TNF-α (r2 = 0.73) when assuming a simple (linear) relationship. CONCLUSION: This study highlights the importance of sample site when investigating the roles of immunoglobulins and cytokines in disease pathogenesis and suggests that both localized and systemic immune responses are at play. The prescription of asthma monoclonal therapy is generally based on systemic evaluation of cytokine and immunoglobulin levels. Our research suggests that this approach may not fully reflect the pathophysiology of the disease and may provide insight into why some patients respond to these targeted therapies while others do not.


Asunto(s)
Asma , Líquido del Lavado Bronquioalveolar , Broncoscopía , Citocinas , Inmunoglobulina E , Humanos , Asma/inmunología , Asma/tratamiento farmacológico , Asma/sangre , Adulto , Masculino , Femenino , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/citología , Persona de Mediana Edad , Citocinas/sangre , Inmunoglobulina E/sangre , Adulto Joven , Inmunoglobulinas/sangre , Anciano
2.
Am J Physiol Gastrointest Liver Physiol ; 323(5): G439-G460, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36165492

RESUMEN

DNA sensor pathways can initiate inflammasome, cell death, and type I interferon (IFN) signaling in immune-mediated inflammatory diseases (IMIDs), including type I interferonopathies. We investigated the involvement of these pathways in the pathogenesis of ulcerative colitis (UC) by analyzing the expression of DNA sensor, inflammasome, and type I IFN biomarker genes in colonic mucosal biopsy tissue from control (n = 31), inactive UC (n = 31), active UC (n = 33), and a UC single-cell RNA-Seq dataset. The effects of type I IFN (IFN-ß), IFN-γ, and TNF-α on gene expression, cytokine production, and cell death were investigated in human colonic organoids. In organoids treated with cytokines alone, or in combination with NLR family pyrin domain-containing 3 (NLRP3), caspase, or JAK inhibitors, cell death was measured, and supernatants were assayed for IL-1ß/IL-18/CXCL10. The expression of DNA sensor pathway genes-PYHIN family members [absent in melanoma 2 (AIM2), IFI16, myeloid cell nuclear differentiation antigen (MNDA), and pyrin and HIN domain family member 1 (PYHIN1)- as well as Z-DNA-binding protein 1 (ZBP1), cyclic GMP-AMP synthase (cGAS), and DDX41 was increased in active UC and expressed in a cell type-restricted pattern. Inflammasome genes (CASP1, IL1B, and IL18), type I IFN inducers [stimulator of interferon response cGAMP interactor 1 (STING), TBK1, and IRF3), IFNB1, and type I IFN biomarker genes (OAS2, IFIT2, and MX2) were also increased in active UC. Cotreatment of organoids with IFN-ß or IFN-γ in combination with TNFα increased expression of IFI16, ZBP1, CASP1, cGAS, and STING induced cell death and IL-1ß/IL-18 secretion. This inflammatory cell death was blocked by the JAK inhibitor tofacitinib but not by inflammasome or caspase inhibitors. Increased type I IFN activity may drive elevated expression of DNA sensor genes and JAK-dependent but inflammasome-independent inflammatory cell death of colonic epithelial cells in UC.NEW & NOTEWORTHY This study found that patients with active UC have significantly increased colonic gene expression of cytosolic DNA sensor, inflammasome, STING, and type I IFN signaling pathways. The type I IFN, IFN-ß, in combination with TNF-α induced JAK-dependent but NLRP3 and inflammasome-independent inflammatory cell death of colonic organoids. This novel inflammatory cell death phenotype is relevant to UC immunopathology and may partially explain the efficacy of the JAKinibs tofacitinib and upadacitinib in patients with UC.


Asunto(s)
Colitis Ulcerosa , Interferón Tipo I , Inhibidores de las Cinasas Janus , Humanos , Inflamasomas/metabolismo , Interleucina-18 , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Necrosis Tumoral alfa , Inhibidores de Caspasas , Organoides/metabolismo , Pirina , Caspasa 1/metabolismo , Nucleotidiltransferasas/metabolismo , ADN , Muerte Celular , Proteínas de Unión al ADN/metabolismo , Antígenos de Diferenciación
3.
Pediatr Allergy Immunol ; 33(12): e13892, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36564884

RESUMEN

Early life dietary patterns and timely maturation of mucosa-associated microbial communities are important factors influencing immune development and for establishing robust immune tolerance networks. Microbial fermentation of dietary components in vivo generates a vast array of molecules, some of which are integral components of the molecular circuitry that regulates immune and metabolic functions. These in turn protect against aberrant inflammatory processes and promote effector immune responses that quickly eliminate pathogens. Multiple studies suggest that changes in dietary habits, altered microbiome composition, and microbial metabolism are associated with asthma risk and disease severity. While it remains unclear whether these microbiome alterations are a cause or consequence of dysregulated immune responses, there is significant potential for using diet in targeted manipulations of the gut microbiome and its metabolic functions in promoting immune health. In this article, we will summarize our knowledge to date on the role of dietary patterns and microbiome activities on immune responses within the airways. Given the malleability of the human microbiome, its integration into the immune system, and its responsiveness to diet, this makes it a highly attractive target for therapeutic and nutritional intervention in children with asthma.


Asunto(s)
Asma , Microbioma Gastrointestinal , Microbiota , Niño , Humanos , Asma/etiología , Dieta , Sistema Inmunológico
4.
Allergy ; 75(5): 1146-1154, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31605638

RESUMEN

BACKGROUND: It is suggested that airway fungi, in particular Aspergillus may impinge on clinical phenotype in asthma. Indeed, the term severe asthma with fungal sensitization (SAFS) has been coined. We aimed to ascertain whether the presence of fungi, in particular Aspergillus fumigatus, in the airway correlated with asthma severity and control. Furthermore, we aimed to determine whether traditional markers of Aspergillus sensitization related to the presence of Aspergillus within the airway. METHODS: Sixty-nine patients characterized by asthma severity (GINA) and level of control (ACQ-7) underwent bronchoscopy and bronchoalveolar lavage (BAL). Serum was assessed for A fumigatus-specific IgE and total IgE. Galactomannan and relevant cytokine levels were assessed in serum, plasma and BAL. BAL was analyzed for the presence of A fumigatus. RESULTS: In BAL, fungi were visible by microscopy in 70% and present by qPCR in 86% of patients, while A fumigatus was detectable by qPCR in 46%. Plasma and BAL IL-4, IL-6, IL-10, IL-13 and TNF-α correlated with BAL fungal presence, while plasma IL-17 correlated with BAL fungal presence. Aspergillus positive BAL correlated with increased plasma and BAL IL-6 and BAL IL-13. There was no relationship between fungal airway presence and steroid dose, asthma severity or control. The presence of Aspergillus within the airway did not relate to serum IgE positivity for Aspergillus. CONCLUSIONS: Fungi were present in a large proportion of our asthmatic patients' airways, but their presence was not predicted by traditional markers of sensitization, nor did it appear to be related to measures of disease severity or control.


Asunto(s)
Aspergilosis Broncopulmonar Alérgica , Asma , Aspergillus fumigatus , Asma/diagnóstico , Líquido del Lavado Bronquioalveolar , Humanos , Inmunoglobulina E , Índice de Severidad de la Enfermedad
5.
Infect Immun ; 87(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31138611

RESUMEN

Urinary tract infections (UTIs) are common and in general are caused by intestinal uropathogenic Escherichia coli (UPEC) ascending via the urethra. Microcompartment-mediated catabolism of ethanolamine, a host cell breakdown product, fuels the competitive overgrowth of intestinal E. coli, both pathogenic enterohemorrhagic E. coli and commensal strains. During a UTI, urease-negative E. coli bacteria thrive, despite the comparative nutrient limitation in urine. The role of ethanolamine as a potential nutrient source during UTIs is understudied. We evaluated the role of the metabolism of ethanolamine as a potential nitrogen and carbon source for UPEC in the urinary tract. We analyzed infected urine samples by culture, high-performance liquid chromatography, reverse transcription-quantitative PCR, and genomic sequencing. The ethanolamine concentration in urine was comparable to the concentration of the most abundant reported urinary amino acid, d-serine. Transcription of the eut operon was detected in the majority of urine samples containing E. coli screened. All sequenced UPEC strains had conserved eut operons, while metabolic genotypes previously associated with UTI (dsdCXA, metE) were mainly limited to phylogroup B2. In vitro ethanolamine was found to be utilized as a sole source of nitrogen by UPEC strains. The metabolism of ethanolamine in artificial urine medium (AUM) induced metabolosome formation and provided a growth advantage at the physiological levels found in urine. Interestingly, eutE (which encodes acetaldehyde dehydrogenase) was required for UPEC strains to utilize ethanolamine to gain a growth advantage in AUM, suggesting that ethanolamine is also utilized as a carbon source. These data suggest that urinary ethanolamine is a significant additional carbon and nitrogen source for infecting E. coli strains.


Asunto(s)
Infecciones por Escherichia coli/metabolismo , Etanolamina/metabolismo , Infecciones Urinarias/metabolismo , Humanos , Operón , Polimorfismo de Nucleótido Simple , Escherichia coli Uropatógena/genética , Escherichia coli Uropatógena/crecimiento & desarrollo
6.
Mol Pharm ; 15(12): 5711-5727, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30388019

RESUMEN

Pharmacokinetic research at the host-microbe interface has been primarily directed toward effects on drug metabolism, with fewer investigations considering the absorption process. We previously demonstrated that the transcriptional expression of genes encoding intestinal transporters involved in lipid translocation are altered in germ-free and conventionalized mice possessing distinct bile acid signatures. It was consequently hypothesized that microbial bile acid metabolism, which is the deconjugation and dehydroxylation of the bile acid steroid nucleus by gut bacteria, may impact upon drug transporter expression and/or activity and potentially alter drug disposition. Using a panel of three human intestinal cell lines (Caco-2, T84, and HT-29) that differ in basal transporter expression level, bile acid conjugation-, and hydroxylation-status was shown to influence the transcription of genes encoding several major influx and efflux transporter proteins. We further investigated if these effects on transporter mRNA would translate to altered drug disposition and activity. The results demonstrated that the conjugation and hydroxylation status of the bile acid steroid nucleus can influence the cellular response to multidrug resistance (MDR) substrates, a finding that did not directly correlate with directionality of gene or protein expression. In particular, we noted that the cytotoxicity of cyclosporine A was significantly augmented in the presence of the unconjugated bile acids deoxycholic acid (DCA) and chenodeoxycholic acid (CDCA) in P-gp positive cell lines, as compared to their taurine/glycine-conjugated counterparts, implicating P-gp in the molecular response. Overall this work identifies a novel mechanism by which gut microbial metabolites may influence drug accumulation and suggests a potential role for the microbial bile acid-deconjugating enzyme bile salt hydrolase (BSH) in ameliorating multidrug resistance through the generation of bile acid species with the capacity to access and inhibit P-gp ATPase. The physicochemical property of nonionization is suggested to underpin the preferential ability of unconjugated bile acids to attenuate the efflux of P-gp substrates and to sensitize tumorigenic cells to cytotoxic therapeutics in vitro. This work provides new impetus to investigate whether perturbation of the gut microbiota, and thereby the bile acid component of the intestinal metabolome, could alter drug pharmacokinetics in vivo. These findings may additionally contribute to the development of less toxic P-gp modulators, which could overcome MDR.


Asunto(s)
Ácido Quenodesoxicólico/metabolismo , Ciclosporina/farmacología , Ácido Desoxicólico/metabolismo , Microbioma Gastrointestinal/fisiología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Variación Biológica Poblacional , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos/fisiología , Glicina , Células HT29 , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , ARN Mensajero/metabolismo , Taurina/metabolismo , Pruebas de Toxicidad
7.
Stem Cells ; 34(5): 1354-68, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26840832

RESUMEN

Disorders affecting smooth muscle structure/function may require technologies that can generate large scale, differentiated and contractile smooth muscle cells (SMC) suitable for cell therapy. To date no clonal precursor population that provides large numbers of differentiated SMC in culture has been identified in a rodent. Identification of such cells may also enhance insight into progenitor cell fate decisions and the relationship between smooth muscle precursors and disease states that implicate differentiated SMC. In this study, we used classic clonal expansion techniques to identify novel self-renewing Islet 1 (Isl-1) positive primitive progenitor cells (PPC) within rat bone marrow that exhibited canonical stem cell markers and preferential differentiation towards a smooth muscle-like fate. We subsequently used molecular tagging to select Isl-1 positive clonal populations from expanded and de novo marrow cell populations. We refer to these previously undescribed cells as the PPC given its stem cell marker profile, and robust self-renewal capacity. PPC could be directly converted into induced smooth muscle cells (iSMC) using single transcription factor (Kruppel-like factor 4) knockdown or transactivator (myocardin) overexpression in contrast to three control cells (HEK 293, endothelial cells and mesenchymal stem cells) where such induction was not possible. iSMC exhibited immuno- and cytoskeletal-phenotype, calcium signaling profile and contractile responses similar to bona fide SMC. Passaged iSMC could be expanded to a scale sufficient for large scale tissue replacement. PPC and reprogramed iSMC so derived may offer future opportunities to investigate molecular, structure/function and cell-based replacement therapy approaches to diverse cardiovascular, respiratory, gastrointestinal, and genitourinary diseases that have as their basis smooth muscle cell functional aberrancy or numerical loss. Stem Cells 2016;34:1354-1368.


Asunto(s)
Reprogramación Celular , Proteínas con Homeodominio LIM/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Miocitos del Músculo Liso/citología , Factores de Transcripción/metabolismo , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Proliferación Celular , Autorrenovación de las Células , Separación Celular , Células Cultivadas , Células Clonales , Silenciador del Gen , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Fenotipo , Ratas Endogámicas F344 , Telomerasa/metabolismo , Transactivadores/metabolismo
8.
Proc Natl Acad Sci U S A ; 111(20): 7421-6, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24799697

RESUMEN

Alterations in the gastrointestinal microbiota have been implicated in obesity in mice and humans, but the key microbial functions influencing host energy metabolism and adiposity remain to be determined. Despite an increased understanding of the genetic content of the gastrointestinal microbiome, functional analyses of common microbial gene sets are required. We established a controlled expression system for the parallel functional analysis of microbial alleles in the murine gut. Using this approach we show that bacterial bile salt hydrolase (BSH) mediates a microbe-host dialogue that functionally regulates host lipid metabolism and plays a profound role in cholesterol metabolism and weight gain in the host. Expression of cloned BSH enzymes in the gastrointestinal tract of gnotobiotic or conventionally raised mice significantly altered plasma bile acid signatures and regulated transcription of key genes involved in lipid metabolism (Pparγ, Angptl4), cholesterol metabolism (Abcg5/8), gastrointestinal homeostasis (RegIIIγ), and circadian rhythm (Dbp, Per1/2) in the liver or small intestine. High-level expression of BSH in conventionally raised mice resulted in a significant reduction in host weight gain, plasma cholesterol, and liver triglycerides, demonstrating the overall impact of elevated BSH activity on host physiology. In addition, BSH activity in vivo varied according to BSH allele group, indicating that subtle differences in activity can have significant effects on the host. In summary, we demonstrate that bacterial BSH activity significantly impacts the systemic metabolic processes and adiposity in the host and represents a key mechanistic target for the control of obesity and hypercholesterolemia.


Asunto(s)
Ácidos y Sales Biliares/química , Tracto Gastrointestinal/microbiología , Metabolismo de los Lípidos/genética , Aumento de Peso/genética , Adiponectina/metabolismo , Adiposidad , Animales , Ritmo Circadiano , Escherichia coli/genética , Vida Libre de Gérmenes , Hidrólisis , Lactobacillus/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Transcripción Genética
9.
Respir Res ; 17(1): 163, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27919249

RESUMEN

Asthma is a chronic respiratory disease whose prevalence is increasing in the western world. Recently research has begun to focus on the role the microbiome plays in asthma pathogenesis in the hope of further understanding this respiratory disorder. Considered sterile until recently, the lungs have revealed themselves to contain a unique microbiota. A shift towards molecular methods for the quantification and sequencing of microbial DNA has revealed that the airways harbour a unique microbiota with apparent, reproducible differences present between healthy and diseased lungs. There is a hope that in classifying the microbial load of the asthmatic airway an insight may be afforded as to the possible role pulmonary microbes may have in propagating an asthmatic airway response. This could potentially pave the way for new therapeutic strategies for the treatment of chronic lung conditions such as asthma.


Asunto(s)
Asma/microbiología , Asma/virología , Pulmón/microbiología , Pulmón/virología , Microbiota , Animales , Asma/epidemiología , Asma/fisiopatología , Carga Bacteriana , Interacciones Huésped-Patógeno , Humanos , Hipótesis de la Higiene , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/fisiopatología , Factores de Riesgo , Transducción de Señal , Carga Viral
11.
Mol Cell Neurosci ; 69: 41-53, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26476273

RESUMEN

The p75 neurotrophin receptor (p75(NTR)) undergoes γ-secretase-mediated regulated intramembrane proteolysis and is involved in glioblastoma cell migration and invasion. Consistent with previous reports, in this study we show that p75NTR increases U87-MG glioblastoma cell migration, which is reversed by inhibition of γ-secretase activity. However, we show that expression or stabilization of the γ-secretase-generated p75(NTR) intracellular domain (ICD) is not sufficient to induce U87-MG glioblastoma cell migration, and that exogenous expression of p75(NTR) ICD inhibits p75(NTR)-mediated glioblastoma cell (U87-MG and U373-MG) migration. To identify pathways and to determine how p75(NTR) mediates glioblastoma migration we utilized a microarray approach to assess differential gene expression profiles between parental U87-MG and cells stably expressing wild-type p75(NTR), a γ-secretase cleavage-resistant chimeric p75(NTR) mutant (p75FasTM) and the γ-secretase-generated p75(NTR)-ICD, which mimics constitutively cleaved p75(NTR) receptor. In our microarray data analysis we identified a subset of genes that were constitutively up-regulated in wild-type p75(NTR) cells, which were also repressed in p75(NTR) ICD expressing cells. Furthermore, our data revealed among the many differentially expressed genes, cadherin-11 (Cdh-11), matrix metalloproteinase 12 and relaxin/insulin-like family peptide receptor 2 as constitutively up-regulated in wild-type p75(NTR) cells, independent of γ-secretase activity. Consistent with a role in glioblastoma migration, we found that U87-p75(NTR) cells express higher levels of Cdh-11 protein and that siRNA-mediated knockdown of Cdh-11 resulted in a significant decrease in p75(NTR)-mediated glioblastoma cell migration. Therefore, we hypothesize that p75(NTR) can impact U87-MG glioblastoma cell migration in a γ-secretase-independent manner through modulation of specific genes, including Cdh-11, and that both γ-secretase-independent and -dependent mechanisms are involved in p75(NTR)-mediated U87-MG glioblastoma cell migration.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Cadherinas/metabolismo , Movimiento Celular/fisiología , Glioblastoma/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Línea Celular , Glioblastoma/patología , Humanos
12.
J Immunol ; 191(12): 6084-92, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24198280

RESUMEN

Listeria monocytogenes is a Gram-positive bacterium that can cause septicemia and meningitis. TLRs are central receptors of the innate immune system that drive inflammatory responses to invading microbes such as L. monocytogenes. Although intestinal epithelial cells (IECs) represent the initial point of entry used by L. monocytogenes for infection, the innate immune response to L. monocytogenes in these cells has been poorly characterized to date. The aim of this study was to determine which TLRs are involved in mediating the immune response to L. monocytogenes in IECs. We performed an RNA interference screen of TLRs 1-10 in the HT-29 IEC cell line and observed the most significant reduction in chemokine output following silencing of TLR10. This effect was also observed in the macrophage cell line THP-1. The chemokines CCL20, CCL1, and IL-8 were reduced following knockdown of TLR10. Silencing of TLR10 resulted in increased viability of L. monocytogenes in both HT-29 and THP-1 cells. TLR10 was found to be predominantly expressed intracellularly in epithelia, and activation required viable L. monocytogenes. NF-κB activation was seen to require TLR2 in addition to TLR10. Taken together, these data indicate novel roles for TLR10 in sensing pathogenic infection in both the epithelium and macrophages and have identified L. monocytogenes as a source of ligand for the orphan receptor TLR10.


Asunto(s)
Células Epiteliales/inmunología , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Listeria monocytogenes/inmunología , Macrófagos/inmunología , Receptor Toll-Like 10/fisiología , Quimiocinas/biosíntesis , Quimiocinas/genética , Células Epiteliales/microbiología , Regulación de la Expresión Génica/inmunología , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Inmunidad Innata , Técnicas In Vitro , Interleucinas/biosíntesis , Interleucinas/genética , Mucosa Intestinal/citología , Ligandos , Macrófagos/microbiología , FN-kappa B/metabolismo , Especificidad de Órganos , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Receptor Toll-Like 10/antagonistas & inhibidores , Receptor Toll-Like 10/genética , Receptor Toll-Like 10/inmunología , Receptor Toll-Like 2/fisiología , Receptores Toll-Like/biosíntesis , Receptores Toll-Like/genética
13.
Proc Natl Acad Sci U S A ; 109(6): 2108-13, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22308390

RESUMEN

Bifidobacteria comprise a significant proportion of the human gut microbiota. Several bifidobacterial strains are currently used as therapeutic interventions, claiming various health benefits by acting as probiotics. However, the precise mechanisms by which they maintain habitation within their host and consequently provide these benefits are not fully understood. Here we show that Bifidobacterium breve UCC2003 produces a cell surface-associated exopolysaccharide (EPS), the biosynthesis of which is directed by either half of a bidirectional gene cluster, thus leading to production of one of two possible EPSs. Alternate transcription of the two opposing halves of this cluster appears to be the result of promoter reorientation. Surface EPS provided stress tolerance and promoted in vivo persistence, but not initial colonization. Marked differences were observed in host immune response: strains producing surface EPS (EPS(+)) failed to elicit a strong immune response compared with EPS-deficient variants. Specifically, EPS production was shown to be linked to the evasion of adaptive B-cell responses. Furthermore, presence of EPS(+) B. breve reduced colonization levels of the gut pathogen Citrobacter rodentium. Our data thus assigns a pivotal and beneficial role for EPS in modulating various aspects of bifidobacterial-host interaction, including the ability of commensal bacteria to remain immunologically silent and in turn provide pathogen protection. This finding enforces the probiotic concept and provides mechanistic insights into health-promoting benefits for both animal and human hosts.


Asunto(s)
Bifidobacterium/inmunología , Membrana Celular/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad/inmunología , Polisacáridos Bacterianos/inmunología , Ácidos , Animales , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Infecciones por Bifidobacteriales/inmunología , Infecciones por Bifidobacteriales/microbiología , Bifidobacterium/crecimiento & desarrollo , Bilis , Citrobacter/crecimiento & desarrollo , Recuento de Colonia Microbiana , Citocinas/metabolismo , Sistema Digestivo/microbiología , Sitios Genéticos/genética , Humanos , Evasión Inmune/inmunología , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/inmunología , Bazo/microbiología
14.
Immunol Cell Biol ; 92(7): 563-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24732075

RESUMEN

The pathogenic gram-positive bacteria, Listeria monocytogenes is a facultative infectious intracellular pathogen that causes listeriosis. Effective elimination of infection is dependent upon a functioning innate immune system and activation of inflammatory responses by pathogen recognition receptors (PRRs). In this review, we trace the route of L. monocytogenes invasion as it disseminates from the intestinal epithelium, through the bloodstream of the host, to the liver and spleen. Along this route, we highlight the diverse, region specific, innate defences in place throughout the course of infection. We provide an overview of recent advances in our knowledge of key innate immune defences against L. monocytogenes, focusing on the PRRs in various cell types known to be critical in the detection of this pathogen.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Inflamasomas/metabolismo , Interferones/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/microbiología , Listeriosis/metabolismo , Listeriosis/transmisión , Receptores de Reconocimiento de Patrones/metabolismo , Receptores Toll-Like/metabolismo
15.
Microbiol Mol Biol Rev ; 88(1): e0009423, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38206006

RESUMEN

SUMMARYHuman milk oligosaccharides (HMOs) are complex, multi-functional glycans present in human breast milk. They represent an intricate mix of heterogeneous structures which reach the infant intestine in an intact form as they resist gastrointestinal digestion. Therefore, they confer a multitude of benefits, directly and/or indirectly, to the developing neonate. Certain bifidobacterial species, being among the earliest gut colonizers of breast-fed infants, have an adapted functional capacity to metabolize various HMO structures. This ability is typically observed in infant-associated bifidobacteria, as opposed to bifidobacteria associated with a mature microbiota. In recent years, information has been gleaned regarding how these infant-associated bifidobacteria as well as certain other taxa are able to assimilate HMOs, including the mechanistic strategies enabling their acquisition and consumption. Additionally, complex metabolic interactions occur between microbes facilitated by HMOs, including the utilization of breakdown products released from HMO degradation. Interest in HMO-mediated changes in microbial composition and function has been the focal point of numerous studies, in recent times fueled by the availability of individual biosynthetic HMOs, some of which are now commonly included in infant formula. In this review, we outline the main HMO assimilatory and catabolic strategies employed by infant-associated bifidobacteria, discuss other taxa that exhibit breast milk glycan degradation capacity, and cover HMO-supported cross-feeding interactions and related metabolites that have been described thus far.


Asunto(s)
Microbioma Gastrointestinal , Leche Humana , Recién Nacido , Femenino , Humanos , Leche Humana/química , Bifidobacterium , Lactancia Materna , Oligosacáridos/análisis , Oligosacáridos/metabolismo
16.
Allergy Asthma Immunol Res ; 15(3): 406-411, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37075793

RESUMEN

The lungs have their own microbiota which seems to be altered in disease processes such as asthma. Viral infection accounts for many asthma exacerbations. Little is known about the lung virome, and the role that viruses play in non-exacerbating asthmatics. We aimed to assess if detection of virus in bronchoscopy samples of asthmatic patients in a non-exacerbating state influences their asthma control and modulates airway cytokine composition. Patients were recruited from a specialist asthma clinic and underwent bronchoscopy with standardised bronchoalveolar lavage (BAL). Viral analysis was performed; cell differential and cytokine levels were measured. Forty-six samples were obtained of which 10.8% demonstrated evidence of airway virus, and 91.3% of patients in the cohort were classed as severe asthmatics. Oral steroid use was significantly higher in severe asthmatic patients with virus detected, and the forced expiratory volume in one second tended to be lower in the virus-detected group. It was also found that BAL interleukin-13 and tumor necrosis factor-α levels were significantly higher in severe asthmatic patients with virus detected. Our results suggest that in severe asthmatics in a non-exacerbating state, the presence of virus resulted in overall poorer asthma control. The pattern of cytokine elevation seen in asthmatic patients with virus detected may provide insight to the pathophysiology involved.

17.
Microbiome ; 11(1): 168, 2023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37528457

RESUMEN

BACKGROUND: Typically, animal models studying gastrointestinal microbiotas compromised in early life have employed either germ-free animals or mice treated with a cocktail of antibiotics. Such studies intend to mimic scenarios of infants born by caesarean section and/or subjected to antibiotic treatment. However, the antibiotics used in these studies are rarely prescribed to infants. Therefore, an early life model was developed in which the murine gastrointestinal microbiota was severely disrupted by clindamycin treatment. RESULTS: In this mouse model, we investigated the extent supplementation with a synbiotic mixture of prebiotics, being scGOS/lcFOS with the human milk oligosaccharide 2'-Fucosyllactose (2'-FL), in combination with or without single strain or mix of "infant type" bifidobacteria, can rescue an antibiotic-compromised microbiota. Shotgun metagenomic sequencing showed that the microbiota was severely disrupted by the clindamycin challenge. No recovery was observed 3 weeks post-challenge in the scGOS/lcFOS/2'FL group, while the group that received the synbiotic treatment of scGOS/lcFOS/2'-FL with Bifidobacterium breve NRBB01 showed partial recovery. Strikingly in the scGOS/lcFOS/2'-FL group receiving the mixture of bifidobacteria resulted in a recovery of the microbiota disruption. Histological analyses showed that the clindamycin-treated animals at the end of the experiment still suffered from mild oedema and villi/colonic crypt irregularities which was ameliorated by the synbiotic intervention. CONCLUSION: Our study demonstrates that supplementation of synbiotic mixture of scGOS/lcFOS/2'-FL in combination with a specific mix of infant-type bifidobacterial strains is able to partially revive an antibiotic-perturbed gastrointestinal microbiota. Video Abstract.


Asunto(s)
Microbioma Gastrointestinal , Simbióticos , Humanos , Lactante , Animales , Embarazo , Ratones , Femenino , Bifidobacterium , Antibacterianos/farmacología , Cesárea , Clindamicina , Oligosacáridos
18.
Sci Rep ; 13(1): 15782, 2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37737287

RESUMEN

As the COVID-19 pandemic winds down, it leaves behind the serious concern that future, even more disruptive pandemics may eventually surface. One of the crucial steps in handling the SARS-CoV-2 pandemic was being able to detect the presence of the virus in an accurate and timely manner, to then develop policies counteracting the spread. Nevertheless, as the pandemic evolved, new variants with potentially dangerous mutations appeared. Faced by these developments, it becomes clear that there is a need for fast and reliable techniques to create highly specific molecular tests, able to uniquely identify VOCs. Using an automated pipeline built around evolutionary algorithms, we designed primer sets for SARS-CoV-2 (main lineage) and for VOC, B.1.1.7 (Alpha) and B.1.1.529 (Omicron). Starting from sequences openly available in the GISAID repository, our pipeline was able to deliver the primer sets for the main lineage and each variant in a matter of hours. Preliminary in-silico validation showed that the sequences in the primer sets featured high accuracy. A pilot test in a laboratory setting confirmed the results: the developed primers were favorably compared against existing commercial versions for the main lineage, and the specific versions for the VOCs B.1.1.7 and B.1.1.529 were clinically tested successfully.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/diagnóstico , Pandemias , Inteligencia Artificial
19.
Infect Immun ; 80(8): 2712-23, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22615252

RESUMEN

The sphingosine-1-phosphate (S1P) analogue FTY720 is therapeutically efficacious in multiple sclerosis and in the prevention of transplant rejection. It prevents the migration of lymphocytes to sites of pathology by trapping them within the peripheral lymph nodes, mesenteric lymph nodes (MLNs), and Peyer's patches. However, evidence suggests that its clinical use may increase the risk of mucosal infections. We investigated the impact of FTY720 treatment on susceptibility to gastrointestinal infection with the mouse enteric pathogen Citrobacter rodentium. This attaching and effacing bacterium induces a transient bacterial colitis in immunocompetent mice that resembles human infection with pathogenic Escherichia coli. FTY720 treatment induced peripheral blood lymphopenia, trapped lymphocytes in the MLNs, and prevented the clearance of bacteria when mice were infected with luciferase-tagged C. rodentium. FTY720-treated C. rodentium-infected mice had enhanced colonic inflammation, with significantly higher colon mass, colon histopathology, and neutrophil infiltration than vehicle-infected animals. In addition, FTY720-treated infected mice had significantly lower numbers of colonic dendritic cells, macrophages, and T cells. Gene expression analysis demonstrated that FTY720-treated infected mice had an impaired innate immune response and a blunted mucosal adaptive immune response, including Th1 cytokines. The data demonstrate that the S1P analogue FTY720 adversely affects the immune response to and clearance of C. rodentium.


Asunto(s)
Citrobacter rodentium/efectos de los fármacos , Infecciones por Enterobacteriaceae/microbiología , Inmunosupresores/farmacología , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Animales , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Femenino , Clorhidrato de Fingolimod , Regulación de la Expresión Génica/inmunología , Inmunidad Mucosa/efectos de los fármacos , Linfopenia/inducido químicamente , Ratones , Ratones Endogámicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Organismos Libres de Patógenos Específicos , Esfingosina/farmacología
20.
Immunology ; 136(3): 312-24, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22385384

RESUMEN

Different rates of bacterial translocation across the gut mucosa have been reported but few studies have examined translocation of commensals at the level of the gut epithelial microfold (M) cell. We used an in vitro M-cell model to quantify translocation and determine the transcriptional response of M cells to various commensal bacteria. The transport kinetics and gene expression profile of M cells in response to different bacterial strains, namely Lactobacillus salivarius, Escherichia coli and Bacteroides fragilis, was assessed. Bacterial strains translocated across M cells with different efficiencies; E. coli and B. fragilis translocated with equal efficiency whereas L. salivarius translocated with less efficiency. Microarray analysis of the M cell response showed both common and differential gene expression changes between the bacterial strains. In the presence of bacteria, but not control beads, up-regulated genes were mainly involved in transcription regulation whereas pro-inflammatory and stress response genes were primarily up-regulated by E. coli and B. fragilis, but not L. salivarius nor beads. Translocation of bacteria and M-cell gene expression responses were confirmed in murine M cells following bacterial challenge in vivo. These results demonstrate that M cells have the ability to discriminate between different commensal bacteria and modify subsequent immune responses.


Asunto(s)
Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Bacteroides fragilis/inmunología , Transporte Biológico Activo/inmunología , Células CACO-2 , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Escherichia coli/inmunología , Femenino , Perfilación de la Expresión Génica , Humanos , Mucosa Intestinal/citología , Lactobacillus/inmunología , Metagenoma/inmunología , Ratones , Ratones Endogámicos BALB C , Modelos Inmunológicos , Monocitos/inmunología , Monocitos/microbiología , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/microbiología , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA