Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 179(2): 417-431.e19, 2019 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585081

RESUMEN

Severe asthma patients with low type 2 inflammation derive less clinical benefit from therapies targeting type 2 cytokines and represent an unmet need. We show that mast cell tryptase is elevated in severe asthma patients independent of type 2 biomarker status. Active ß-tryptase allele count correlates with blood tryptase levels, and asthma patients carrying more active alleles benefit less from anti-IgE treatment. We generated a noncompetitive inhibitory antibody against human ß-tryptase, which dissociates active tetramers into inactive monomers. A 2.15 Å crystal structure of a ß-tryptase/antibody complex coupled with biochemical studies reveal the molecular basis for allosteric destabilization of small and large interfaces required for tetramerization. This anti-tryptase antibody potently blocks tryptase enzymatic activity in a humanized mouse model, reducing IgE-mediated systemic anaphylaxis, and inhibits airway tryptase in Ascaris-sensitized cynomolgus monkeys with favorable pharmacokinetics. These data provide a foundation for developing anti-tryptase as a clinical therapy for severe asthma.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Asma/terapia , Mastocitos/enzimología , Mastocitos/inmunología , Triptasas/antagonistas & inhibidores , Triptasas/inmunología , Adolescente , Regulación Alostérica/inmunología , Animales , Línea Celular , Femenino , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Conejos
3.
Proc Natl Acad Sci U S A ; 119(38): e2123117119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36099298

RESUMEN

Acinetobacter baumannii is a clinically important, predominantly health care-associated gram-negative bacterium with high rates of emerging resistance worldwide. Given the urgent need for novel antibacterial therapies against A. baumannii, we focused on inhibiting lipoprotein biosynthesis, a pathway that is essential for envelope biogenesis in gram-negative bacteria. The natural product globomycin, which inhibits the essential type II signal peptidase prolipoprotein signal peptidase (LspA), is ineffective against wild-type A. baumannii clinical isolates due to its poor penetration through the outer membrane. Here, we describe a globomycin analog, G5132, that is more potent against wild-type and clinical A. baumannii isolates. Mutations leading to G5132 resistance in A. baumannii map to the signal peptide of a single hypothetical gene, which we confirm encodes an alanine-rich lipoprotein and have renamed lirL (prolipoprotein signal peptidase inhibitor resistance lipoprotein). LirL is a highly abundant lipoprotein primarily localized to the inner membrane. Deletion of lirL leads to G5132 resistance, inefficient cell division, increased sensitivity to serum, and attenuated virulence. Signal peptide mutations that confer resistance to G5132 lead to the accumulation of diacylglyceryl-modified LirL prolipoprotein in untreated cells without significant loss in cell viability, suggesting that these mutations overcome a block in lipoprotein biosynthetic flux by decreasing LirL prolipoprotein substrate sensitivity to processing by LspA. This study characterizes a lipoprotein that plays a critical role in resistance to LspA inhibitors and validates lipoprotein biosynthesis as a antibacterial target in A. baumannii.


Asunto(s)
Acinetobacter baumannii , Antibacterianos , Ácido Aspártico Endopeptidasas , Proteínas Bacterianas , Farmacorresistencia Bacteriana , Furanos , Eliminación de Gen , Lipoproteínas , Inhibidores de Proteasas , Piridinas , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/enzimología , Acinetobacter baumannii/genética , Antibacterianos/farmacología , Ácido Aspártico Endopeptidasas/genética , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Furanos/farmacología , Lipoproteínas/biosíntesis , Lipoproteínas/genética , Péptidos/farmacología , Inhibidores de Proteasas/farmacología , Señales de Clasificación de Proteína/genética , Piridinas/farmacología
4.
Nat Immunol ; 13(4): 396-404, 2012 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-22366892

RESUMEN

Immunoglobulin E (IgE) antibodies are pathogenic in asthma and allergic diseases, but the in vivo biology of IgE-producing (IgE(+)) cells is poorly understood. A model of the differentiation of IgE(+) B cells proposes that IgE(+) cells develop through a germinal-center IgG1(+) intermediate and that IgE memory resides in the compartment of IgG1(+) memory B cells. Here we have used a reporter mouse expressing green fluorescent protein associated with membrane IgE transcripts (IgE-GFP) to assess in vivo IgE responses. In contrast to the IgG1-centered model of IgE switching and memory, we found that IgE(+) cells developed through a germinal-center IgE(+) intermediate to form IgE(+) memory B cells and plasma cells. Our studies delineate a new model for the in vivo biology of IgE switching and memory.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular/inmunología , Centro Germinal/citología , Inmunoglobulina E/inmunología , Memoria Inmunológica/inmunología , Células Plasmáticas/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Sustitución del Gen , Centro Germinal/inmunología , Humanos , Cambio de Clase de Inmunoglobulina/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Células Plasmáticas/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Allergy ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38686450

RESUMEN

BACKGROUND: The effects of inhaled corticosteroids (ICS) on healthy airways are poorly defined. OBJECTIVES: To delineate the effects of ICS on gene expression in healthy airways, without confounding caused by changes in disease-related genes and disease-related alterations in ICS responsiveness. METHODS: Randomized open-label bronchoscopy study of high-dose ICS therapy in 30 healthy adult volunteers randomized 2:1 to (i) fluticasone propionate 500 mcg bd daily or (ii) no treatment, for 4 weeks. Laboratory staff were blinded to allocation. Biopsies and brushings were analysed by immunohistochemistry, bulk RNA sequencing, DNA methylation array and metagenomics. RESULTS: ICS induced small between-group differences in blood and lamina propria eosinophil numbers, but not in other immunopathological features, blood neutrophils, FeNO, FEV1, microbiome or DNA methylation. ICS treatment upregulated 72 genes in brushings and 53 genes in biopsies, and downregulated 82 genes in brushings and 416 genes in biopsies. The most downregulated genes in both tissues were canonical markers of type-2 inflammation (FCER1A, CPA3, IL33, CLEC10A, SERPINB10 and CCR5), T cell-mediated adaptive immunity (TARP, TRBC1, TRBC2, PTPN22, TRAC, CD2, CD8A, HLA-DQB2, CD96, PTPN7), B-cell immunity (CD20, immunoglobulin heavy and light chains) and innate immunity, including CD48, Hobit, RANTES, Langerin and GFI1. An IL-17-dependent gene signature was not upregulated by ICS. CONCLUSIONS: In healthy airways, 4-week ICS exposure reduces gene expression related to both innate and adaptive immunity, and reduces markers of type-2 inflammation. This implies that homeostasis in health involves tonic type-2 signalling in the airway mucosa, which is exquisitely sensitive to ICS.

6.
Nature ; 557(7704): 196-201, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29720648

RESUMEN

The movement of core-lipopolysaccharide across the inner membrane of Gram-negative bacteria is catalysed by an essential ATP-binding cassette transporter, MsbA. Recent structures of MsbA and related transporters have provided insights into the molecular basis of active lipid transport; however, structural information about their pharmacological modulation remains limited. Here we report the 2.9 Å resolution structure of MsbA in complex with G907, a selective small-molecule antagonist with bactericidal activity, revealing an unprecedented mechanism of ABC transporter inhibition. G907 traps MsbA in an inward-facing, lipopolysaccharide-bound conformation by wedging into an architecturally conserved transmembrane pocket. A second allosteric mechanism of antagonism occurs through structural and functional uncoupling of the nucleotide-binding domains. This study establishes a framework for the selective modulation of ABC transporters and provides rational avenues for the design of new antibiotics and other therapeutics targeting this protein family.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/química , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Quinolinas/química , Quinolinas/farmacología , Transportadoras de Casetes de Unión a ATP/metabolismo , Regulación Alostérica/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Escherichia coli/química , Hidrocarburos/química , Hidrocarburos/metabolismo , Lipopolisacáridos/química , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Modelos Moleculares , Dominios Proteicos/efectos de los fármacos
7.
Allergy ; 77(10): 2974-2986, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35579040

RESUMEN

BACKGROUND: The most recognizable phenotype of severe asthma comprises people who are blood eosinophil and FeNO-high, driven by type 2 (T2) cytokine biology, which responds to targeted biological therapies. However, in many people with severe asthma, these T2 biomarkers are suppressed but poorly controlled asthma persists. The mechanisms driving asthma in the absence of T2 biology are poorly understood. OBJECTIVES: To explore airway pathology in T2 biomarker-high and -low severe asthma. METHODS: T2 biomarker-high severe asthma (T2-high, n = 17) was compared with biomarker-intermediate (T2-intermediate, n = 21) and biomarker-low (T2-low, n = 20) severe asthma and healthy controls (n = 28). Bronchoscopy samples were processed for immunohistochemistry, and sputum for cytokines, PGD2 and LTE4 measurements. RESULTS: Tissue eosinophil, neutrophil and mast cell counts were similar across severe asthma phenotypes and not increased when compared to healthy controls. In contrast, the remodelling features of airway smooth muscle mass and MUC5AC expression were increased in all asthma groups compared with health, but similar across asthma subgroups. Submucosal glands were increased in T2-intermediate and T2-low asthma. In spite of similar tissue cellular inflammation, sputum IL-4, IL-5 and CCL26 were increased in T2-high versus T2-low asthma, and several further T2-associated cytokines, PGD2 and LTE4 , were increased in T2-high and T2-intermediate asthma compared with healthy controls. CONCLUSIONS: Eosinophilic tissue inflammation within proximal airways is suppressed in T2 biomarker-high and T2-low severe asthma, but inflammatory and structural cell activation is present, with sputum T2-associated cytokines highest in T2 biomarker-high patients. Airway remodelling persists and may be important for residual disease expression beyond eosinophilic exacerbations. Registered at ClincialTrials.gov: NCT02883530.


Asunto(s)
Asma , Eosinofilia , Remodelación de las Vías Aéreas (Respiratorias) , Asma/metabolismo , Biomarcadores , Citocinas/análisis , Eosinofilia/patología , Eosinófilos/metabolismo , Humanos , Inflamación/patología , Interleucina-4 , Interleucina-5/análisis , Esputo
8.
J Bacteriol ; 203(13): e0014921, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-33875545

RESUMEN

Lipoprotein diacylglyceryl transferase (Lgt) catalyzes the first step in the biogenesis of Gram-negative bacterial lipoproteins which play crucial roles in bacterial growth and pathogenesis. We demonstrate that Lgt depletion in a clinical uropathogenic Escherichia coli strain leads to permeabilization of the outer membrane and increased sensitivity to serum killing and antibiotics. Importantly, we identify G2824 as the first-described Lgt inhibitor that potently inhibits Lgt biochemical activity in vitro and is bactericidal against wild-type Acinetobacter baumannii and E. coli strains. While deletion of a gene encoding a major outer membrane lipoprotein, lpp, leads to rescue of bacterial growth after genetic depletion or pharmacologic inhibition of the downstream type II signal peptidase, LspA, no such rescue of growth is detected after Lgt depletion or treatment with G2824. Inhibition of Lgt does not lead to significant accumulation of peptidoglycan-linked Lpp in the inner membrane. Our data validate Lgt as a novel antibacterial target and suggest that, unlike downstream steps in lipoprotein biosynthesis and transport, inhibition of Lgt may not be sensitive to one of the most common resistance mechanisms that invalidate inhibitors of bacterial lipoprotein biosynthesis and transport. IMPORTANCE As the emerging threat of multidrug-resistant (MDR) bacteria continues to increase, no new classes of antibiotics have been discovered in the last 50 years. While previous attempts to inhibit the lipoprotein biosynthetic (LspA) or transport (LolCDE) pathways have been made, most efforts have been hindered by the emergence of a common mechanism leading to resistance, namely, the deletion of the gene encoding a major Gram-negative outer membrane lipoprotein lpp. Our unexpected finding that inhibition of Lgt is not susceptible to lpp deletion-mediated resistance uncovers the complexity of bacterial lipoprotein biogenesis and the corresponding enzymes involved in this essential outer membrane biogenesis pathway and potentially points to new antibacterial targets in this pathway.


Asunto(s)
Escherichia coli/metabolismo , Lipoproteínas/metabolismo , Transferasas/metabolismo , Animales , Antibacterianos/farmacología , Ácido Aspártico Endopeptidasas , Proteínas Bacterianas , Escherichia coli/genética , Femenino , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Ratones , Peptidoglicano/metabolismo , Transferasas/química , Transferasas/genética , Escherichia coli Uropatógena/genética , Escherichia coli Uropatógena/metabolismo
9.
Nature ; 528(7580): 127-31, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26580007

RESUMEN

Prevailing dogma holds that cell-cell communication through Notch ligands and receptors determines binary cell fate decisions during progenitor cell divisions, with differentiated lineages remaining fixed. Mucociliary clearance in mammalian respiratory airways depends on secretory cells (club and goblet) and ciliated cells to produce and transport mucus. During development or repair, the closely related Jagged ligands (JAG1 and JAG2) induce Notch signalling to determine the fate of these lineages as they descend from a common proliferating progenitor. In contrast to such situations in which cell fate decisions are made in rapidly dividing populations, cells of the homeostatic adult airway epithelium are long-lived, and little is known about the role of active Notch signalling under such conditions. To disrupt Jagged signalling acutely in adult mammals, here we generate antibody antagonists that selectively target each Jagged paralogue, and determine a crystal structure that explains selectivity. We show that acute Jagged blockade induces a rapid and near-complete loss of club cells, with a concomitant gain in ciliated cells, under homeostatic conditions without increased cell death or division. Fate analyses demonstrate a direct conversion of club cells to ciliated cells without proliferation, meeting a conservative definition of direct transdifferentiation. Jagged inhibition also reversed goblet cell metaplasia in a preclinical asthma model, providing a therapeutic foundation. Our discovery that Jagged antagonism relieves a blockade of cell-to-cell conversion unveils unexpected plasticity, and establishes a model for Notch regulation of transdifferentiation.


Asunto(s)
Anticuerpos/uso terapéutico , Transdiferenciación Celular , Pulmón/citología , Pulmón/metabolismo , Receptores Notch/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Asma/tratamiento farmacológico , Asma/metabolismo , Asma/patología , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/inmunología , Proteínas de Unión al Calcio/metabolismo , Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Rastreo Celular , Transdiferenciación Celular/efectos de los fármacos , Cilios/metabolismo , Modelos Animales de Enfermedad , Femenino , Células Caliciformes/citología , Células Caliciformes/efectos de los fármacos , Células Caliciformes/patología , Homeostasis/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/inmunología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteína Jagged-2 , Ligandos , Pulmón/efectos de los fármacos , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Serrate-Jagged , Transducción de Señal/efectos de los fármacos
10.
Clin Exp Allergy ; 50(12): 1342-1351, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32909660

RESUMEN

BACKGROUND: The anti-interleukin 13 (IL-13) monoclonal antibody lebrikizumab improves lung function in patients with moderate-to-severe uncontrolled asthma, but its effects on airway inflammation and remodelling are unknown. CLAVIER was designed to assess lebrikizumab's effect on eosinophilic inflammation and remodelling. OBJECTIVE: To report safety and efficacy results from enrolled participants with available data from CLAVIER. METHODS: We performed bronchoscopy on patients with uncontrolled asthma before and after 12 weeks of randomized double-blinded treatment with lebrikizumab (n = 31) or placebo (n = 33). The pre-specified primary end-point was relative change in airway subepithelial eosinophils per mm2 of basement membrane (cells/mm2 ). Pre-specified secondary and exploratory outcomes included change in IL-13-associated biomarkers and measures of airway remodelling. RESULTS: There was a baseline imbalance in tissue eosinophils and high variability between treatment groups. There was no discernible change in adjusted mean subepithelial eosinophils/mm2 in response to lebrikizumab (95% CI, -82.5%, 97.5%). As previously observed, FEV1 increased after lebrikizumab treatment. Moreover, subepithelial collagen thickness decreased 21.5% after lebrikizumab treatment (95% CI, -32.9%, -10.2%), and fractional exhaled nitric oxide, CCL26 and SERPINB2 mRNA expression in bronchial tissues also reduced. Lebrikizumab was well tolerated, with a safety profile consistent with other lebrikizumab asthma studies. CONCLUSIONS & CLINICAL RELEVANCE: We did not observe reduced tissue eosinophil numbers in association with lebrikizumab treatment. However, in pre-specified exploratory analyses, lebrikizumab treatment was associated with reduced degree of subepithelial fibrosis, a feature of airway remodelling, as well as improved lung function and reduced key pharmacodynamic biomarkers in bronchial tissues. These results reinforce the importance of IL-13 in airway pathobiology and suggest that neutralization of IL-13 may reduce asthmatic airway remodelling. CLINICAL TRIAL REGISTRATION: NCT02099656.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Antiasmáticos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Asma/tratamiento farmacológico , Eosinófilos/efectos de los fármacos , Interleucina-13/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Adolescente , Adulto , Anciano , Antiasmáticos/efectos adversos , Antiasmáticos/farmacocinética , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Asma/inmunología , Asma/fisiopatología , Método Doble Ciego , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Humanos , Pulmón/inmunología , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Transducción de Señal , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
11.
Nat Immunol ; 14(12): 1302-4, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24240161
12.
Proc Natl Acad Sci U S A ; 114(42): 11223-11228, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-28973946

RESUMEN

The type VII secretion system (T7SS) of Staphylococcus aureus is a multiprotein complex dedicated to the export of several virulence factors during host infection. This virulence pathway plays a key role in promoting bacterial survival and the long-term persistence of staphylococcal abscess communities. The expression of the T7SS is activated by bacterial interaction with host tissues including blood serum, nasal secretions, and pulmonary surfactant. In this work we identify the major stimulatory factors as host-specific cis-unsaturated fatty acids. Increased T7SS expression requires host fatty acid incorporation into bacterial biosynthetic pathways by the Saureus fatty acid kinase (FAK) complex, and FakA is required for virulence. The incorporated cis-unsaturated fatty acids decrease Saureus membrane fluidity, and these altered membrane dynamics are partially responsible for T7SS activation. These data define a molecular mechanism by which Saureus cells sense the host environment and implement appropriate virulence pathways.


Asunto(s)
Interacciones Huésped-Patógeno , Ácido Linoleico/metabolismo , Staphylococcus aureus/fisiología , Sistemas de Secreción Tipo VII/fisiología , Animales , Humanos , Ratones , Factores de Virulencia/metabolismo
13.
J Immunol ; 199(4): 1238-1249, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28696253

RESUMEN

Ox40 ligand (Ox40L) locus genetic variants are associated with the risk for systemic lupus erythematosus (SLE); however, it is unclear how Ox40L contributes to SLE pathogenesis. In this study, we evaluated the contribution of Ox40L and its cognate receptor, Ox40, using in vivo agonist and antagonist approaches in the NZB × NZW (NZB/W) F1 mouse model of SLE. Ox40 was highly expressed on several CD4 Th cell subsets in the spleen and kidney of diseased mice, and expression correlated with disease severity. Treatment of aged NZB/W F1 mice with agonist anti-Ox40 mAbs potently exacerbated renal disease, which was accompanied by activation of kidney-infiltrating T cells and cytokine production. The agonist mAbs also induced activation and inflammatory gene expression in splenic CD4 T cells, including IFN-regulated genes, increased the number of follicular helper T cells and plasmablasts in the spleen, and led to elevated levels of serum IgM and enhanced renal glomerular IgM deposition. In a type I IFN-accelerated lupus model, treatment with an antagonist Ox40:Fc fusion protein significantly delayed the onset of severe proteinuria and improved survival. These data support the hypothesis that the Ox40/Ox40L pathway drives cellular and humoral autoimmune responses during lupus nephritis in NZB/W F1 mice and emphasize the potential clinical value of targeting this pathway in human lupus.


Asunto(s)
Nefritis Lúpica/inmunología , Glicoproteínas de Membrana/metabolismo , Células Plasmáticas/inmunología , Receptores OX40/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Necrosis Tumoral/metabolismo , Animales , Anticuerpos Antinucleares/inmunología , Anticuerpos Monoclonales/inmunología , Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Riñón/inmunología , Riñón/patología , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Nefritis Lúpica/fisiopatología , Ratones , Ratones Endogámicos NZB , Ligando OX40 , Proteinuria/inmunología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/metabolismo
14.
J Allergy Clin Immunol ; 142(5): 1457-1468, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29550052

RESUMEN

BACKGROUND: Asthma is a complex chronic disease underpinned by pathological changes within the airway wall. How variations in structural airway pathology and cellular inflammation contribute to the expression and severity of asthma are poorly understood. OBJECTIVES: Therefore we evaluated pathological heterogeneity using topological data analysis (TDA) with the aim of visualizing disease clusters and microclusters. METHODS: A discovery population of 202 adult patients (142 asthmatic patients and 60 healthy subjects) and an external replication population (59 patients with severe asthma) were evaluated. Pathology and gene expression were examined in bronchial biopsy samples. TDA was applied by using pathological variables alone to create pathology-driven visual networks. RESULTS: In the discovery cohort TDA identified 4 groups/networks with multiple microclusters/regions of interest that were masked by group-level statistics. Specifically, TDA group 1 consisted of a high proportion of healthy subjects, with a microcluster representing a topological continuum connecting healthy subjects to patients with mild-to-moderate asthma. Three additional TDA groups with moderate-to-severe asthma (Airway Smooth MuscleHigh, Reticular Basement MembraneHigh, and RemodelingLow groups) were identified and contained numerous microclusters with varying pathological and clinical features. Mutually exclusive TH2 and TH17 tissue gene expression signatures were identified in all pathological groups. Discovery and external replication applied to the severe asthma subgroup identified only highly similar "pathological data shapes" through analyses of persistent homology. CONCLUSIONS: We have identified and replicated novel pathological phenotypes of asthma using TDA. Our methodology is applicable to other complex chronic diseases.


Asunto(s)
Asma/patología , Bronquios/patología , Adulto , Remodelación de las Vías Aéreas (Respiratorias) , Asma/genética , Bronquios/metabolismo , Análisis por Conglomerados , Estudios de Cohortes , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
15.
Proc Natl Acad Sci U S A ; 112(47): 14664-9, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26561586

RESUMEN

Dendritic cells (DCs) link innate and adaptive immunity and use a host of innate immune and inflammatory receptors to respond to pathogens and inflammatory stimuli. Although DC maturation via canonical NF-κB signaling is critical for many of these functions, the role of noncanonical NF-κB signaling via the serine/threonine kinase NIK (NF-κB-inducing kinase) remains unclear. Because NIK-deficient mice lack secondary lymphoid organs, we generated transgenic mice with targeted NIK deletion in CD11c(+) cells. Although these mice exhibited normal lymphoid organs, they were defective in cross-priming naive CD8(+) T cells following vaccination, even in the presence of anti-CD40 or polyinosinic:polycytidylic acid to induce DC maturation. This impairment reflected two intrinsic defects observed in splenic CD8(+) DCs in vitro, namely antigen cross-presentation to CD8(+) T cells and secretion of IL-12p40, a cytokine known to promote cross-priming in vivo. In contrast, antigen presentation to CD4(+) T cells was not affected. These findings reveal that NIK, and thus probably the noncanonical NF-κB pathway, is critical to allow DCs to acquire the capacity to cross-present antigen and prime CD8 T cells after exposure to licensing stimuli, such as an agonistic anti-CD40 antibody or Toll-like receptor 3 ligand.


Asunto(s)
Antígenos CD40/metabolismo , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Presentación de Antígeno/inmunología , Antígeno CD11c/metabolismo , Eliminación de Gen , Integrasas/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Ratones Transgénicos , Bazo/citología , Quinasa de Factor Nuclear kappa B
16.
J Immunol ; 195(3): 953-64, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26116508

RESUMEN

NF-κB-inducing kinase (NIK) is a primary regulator of the noncanonical NF-κB signaling pathway, which plays a vital role downstream of BAFF, CD40L, lymphotoxin, and other inflammatory mediators. Germline deletion or inactivation of NIK in mice results in the defective development of B cells and secondary lymphoid organs, but the role of NIK in adult animals has not been studied. To address this, we generated mice containing a conditional allele of NIK. Deletion of NIK in adult mice results in decreases in B cell populations in lymph nodes and spleen, similar to what is observed upon blockade of BAFF. Consistent with this, B cells from mice in which NIK is acutely deleted fail to respond to BAFF stimulation in vitro and in vivo. In addition, mice with induced NIK deletion exhibit a significant decrease in germinal center B cells and serum IgA, which is indicative of roles for NIK in additional pathways beyond BAFF signaling. Our conditional NIK-knockout mice may be broadly useful for assessing the postdevelopmental and cell-specific roles of NIK and the noncanonical NF-κB pathway in mice.


Asunto(s)
Factor Activador de Células B/genética , Linfocitos B/inmunología , Activación de Linfocitos/genética , Subunidad p52 de NF-kappa B/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Animales , Linfocitos B/citología , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Mutación de Línea Germinal , Quinasa I-kappa B/metabolismo , Inmunoglobulina A/sangre , Ganglios Linfáticos/citología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Subunidad p52 de NF-kappa B/genética , Eliminación de Secuencia , Transducción de Señal/genética , Transducción de Señal/inmunología , Bazo/citología , Tamoxifeno/farmacología , Quinasa de Factor Nuclear kappa B
17.
J Bacteriol ; 198(14): 2001-2015, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27161118

RESUMEN

UNLABELLED: De novo guanine biosynthesis is an evolutionarily conserved pathway that creates sufficient nucleotides to support DNA replication, transcription, and translation. Bacteria can also salvage nutrients from the environment to supplement the de novo pathway, but the relative importance of either pathway during Staphylococcus aureus infection is not known. In S. aureus, genes important for both de novo and salvage pathways are regulated by a guanine riboswitch. Bacterial riboswitches have attracted attention as a novel class of antibacterial drug targets because they have high affinity for small molecules, are absent in humans, and regulate the expression of multiple genes, including those essential for cell viability. Genetic and biophysical methods confirm the existence of a bona fide guanine riboswitch upstream of an operon encoding xanthine phosphoribosyltransferase (xpt), xanthine permease (pbuX), inosine-5'-monophosphate dehydrogenase (guaB), and GMP synthetase (guaA) that represses the expression of these genes in response to guanine. We found that S. aureus guaB and guaA are also transcribed independently of riboswitch control by alternative promoter elements. Deletion of xpt-pbuX-guaB-guaA genes resulted in guanine auxotrophy, failure to grow in human serum, profound abnormalities in cell morphology, and avirulence in mouse infection models, whereas deletion of the purine salvage genes xpt-pbuX had none of these effects. Disruption of guaB or guaA recapitulates the xpt-pbuX-guaB-guaA deletion in vivo In total, the data demonstrate that targeting the guanine riboswitch alone is insufficient to treat S. aureus infections but that inhibition of guaA or guaB could have therapeutic utility. IMPORTANCE: De novo guanine biosynthesis and purine salvage genes were reported to be regulated by a guanine riboswitch in Staphylococcus aureus We demonstrate here that this is not true, because alternative promoter elements that uncouple the de novo pathway from riboswitch regulation were identified. We found that in animal models of infection, the purine salvage pathway is insufficient for S. aureus survival in the absence of de novo guanine biosynthesis. These data suggest targeting the de novo guanine biosynthesis pathway may have therapeutic utility in the treatment of S. aureus infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Guanina/biosíntesis , Purinas/metabolismo , Riboswitch , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Animales , Proteínas Bacterianas/genética , Femenino , Humanos , Ratones , Staphylococcus aureus/genética
18.
Proc Natl Acad Sci U S A ; 110(39): 15770-5, 2013 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-24019479

RESUMEN

Preceding antibody constant regions are switch (S) regions varying in length and repeat density that are targets of activation-induced cytidine deaminase. We asked how participating S regions influence each other to orchestrate rearrangements at the IgH locus by engineering mice in which the weakest S region, Sε, is replaced with prominent recombination hotspot Sµ. These mice produce copious polyclonal IgE upon challenge, providing a platform to study IgE biology and therapeutic interventions. The insertion enhances ε germ-line transcript levels, shows a preference for direct vs. sequential switching, and reduces intraswitch recombination events at native Sµ. These results suggest that the sufficiency of Sµ to mediate IgH rearrangements may be influenced by context-dependent cues.


Asunto(s)
Cambio de Clase de Inmunoglobulina/genética , Inmunoglobulina E/metabolismo , Recombinación Genética , Alelos , Animales , Linfocitos B/metabolismo , Técnicas de Sustitución del Gen , Marcación de Gen , Sitios Genéticos/genética , Células Germinativas/metabolismo , Hibridomas , Cadenas epsilon de Inmunoglobulina/genética , Cadenas mu de Inmunoglobulina/genética , Activación de Linfocitos/genética , Ratones , Modelos Animales , ARN Mensajero/genética , ARN Mensajero/metabolismo
19.
J Allergy Clin Immunol ; 129(1): 104-11.e1-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21975173

RESUMEN

BACKGROUND: Thymic stromal lymphopoietin (TSLP) is a cytokine implicated in the pathophysiology of asthma through 2 distinct pathways: a TSLP-OX40 ligand (OX40L)-T cell axis and a TSLP-mast cell axis. Whether these pathways are active in human asthma is unknown. OBJECTIVE: We sought to investigate whether mucosal TSLP protein expression relates to asthma severity and distinct immunologic pathways. METHODS: In healthy subjects and patients with mild-to-severe asthma, we immunostained bronchial biopsy specimens for TSLP, OX40, OX40L, T(H)2 cytokines, and inflammatory cell markers. We examined gene expression using RNA microarrays and quantitative RT-PCR. RESULTS: There was considerable heterogeneity in the levels of TSLP, IL-13, and IL-4 immunostaining across the cohort of asthmatic patients examined. Overall, TSLP protein expression was significantly increased in airway epithelium and lamina propria of asthmatic patients, particularly in patients with severe asthma. TSLP immunostaining in both compartments correlated with the severity of airflow obstruction. The majority of leukocytes expressing IL-13 were possibly nuocytes. Accounting for intersubject variability, the 55% of asthmatic patients with increased IL-13 immunostaining in the lamina propria also had increased IL-4 and TSLP expression. This was further substantiated by significant correlations between TSLP gene expression, a T(H)2 gene expression signature, and eosinophilic inflammation in bronchial biopsy specimens. Immunostaining for OX40, OX40L, and CD83 was sparse, with no difference between asthmatic patients and healthy subjects. CONCLUSION: TSLP expression is increased in a subset of patients with severe asthma in spite of high-dose inhaled or oral corticosteroid therapy. Targeting TSLP might only be efficacious in the subset of asthma characterized by increased TSLP expression and T(H)2 inflammation.


Asunto(s)
Asma/inmunología , Citocinas/metabolismo , Asma/genética , Asma/metabolismo , Citocinas/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Eosinofilia/inmunología , Eosinofilia/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inflamación/inmunología , Inflamación/patología , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ligando OX40/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Linfopoyetina del Estroma Tímico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA