Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mucosal Immunol ; 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39038754

RESUMEN

Mast cells (MCs) are derived from CD34+ hematopoietic progenitors, consist of different subtypes, and are involved in several inflammatory conditions. However, our understanding of human MC developmental trajectories and subtypes has been limited by a scarcity of suitable cellular model systems. Herein, we developed an in vitro model of human MC differentiation from induced pluripotent stem cells (iPSC) to study human MC differentiation trajectories. Flow cytometry characterization of hemopoietic cells derived from the myeloid cells-forming complex (MCFC) revealed an initial increase in Lin- CD34+ hematopoietic progenitors within Weeks 1-3, followed by an increase in CD34- CD45RA- SSClow and SSChigh hematopoietic cells. The Lin- CD34+ hematopoietic progenitors consisted of SSClow CD45RA- CD123± c-Kit+ FcεRI+ populations that were ß7-integrinhigh CD203c+ and ß7-integrinhigh CD203c- cells consistent with CMPFcεRI+ cells. Flow cytometry and cytologic analyses of the CD34- Lin- (SSClow) population revealed hypogranular cell populations, predominantly characterized by CD45RA- CD123± c-Kit+ FcεRI- ß7-integrinlow and CD45RA- CD123± c-Kit- FcεRI+ ß7-integrinMid cells. Analyses of hypergranular SSChigh cells identified Lin- CD34- CD45RA- c-Kit+ FcεRI- and Lin- CD34- CD45RA- c-Kit+ FcεRI+ cells. scRNA-seq analysis of the cells harvested at week 4 of the MCFC culture revealed the presence of monocyte and granulocyte progenitors (n = 547 cells, 26.7 %), Erythrocyte / unknown (n = 85, 4.1 %), neutrophils / myelocytes (n = 211 cells, 10.2 %), mast cell progenitor 1 (n = 599, 29.1 %), mast cell progenitor 2 (n = 152, 7.4 %), committed mast cell precursor (n = 113, 5.5 %), and MCs (n = 353, 17.1 %). In silico analyses of the MC precursor and mature MC populations revealed transcriptionally distinct MC precursor subtype and mature MC states (CMA1+ and CMA1- subtypes). Culturing MC precursor populations in MC maturation media (mast cell media II) led to homogenous mature MC populations as evidenced by high expression of high-affinity IgE receptor, metachromatic granules, presence of MC granule proteins (Tryptase and Chymase) and activation following substance P stimulation and FcεRI crosslinking. This human iPSC-based approach generates MC precursors and phenotypically mature and functional MC populations. This system will be a useful model to generate human MC populations and broaden our understanding of MC biology and transcriptional regulation of MC differentiation trajectories.

2.
J Allergy Clin Immunol ; 150(4): 817-829.e6, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35643377

RESUMEN

BACKGROUND: Asthma and chronic obstructive pulmonary disease (COPD) are common chronic respiratory diseases, and some patients have overlapping disease features, termed asthma-COPD overlap (ACO). Patients characterized with ACO have increased disease severity; however, the mechanisms driving this have not been widely studied. OBJECTIVES: This study sought to characterize the phenotypic and transcriptomic features of experimental ACO in mice induced by chronic house dust mite antigen and cigarette smoke exposure. METHODS: Female BALB/c mice were chronically exposed to house dust mite antigen for 11 weeks to induce experimental asthma, cigarette smoke for 8 weeks to induce experimental COPD, or both concurrently to induce experimental ACO. Lung inflammation, structural changes, and lung function were assessed. RNA-sequencing was performed on separated airway and parenchyma lung tissues to assess transcriptional changes. Validation of a novel upstream driver SPI1 in experimental ACO was assessed using the pharmacological SPI1 inhibitor, DB2313. RESULTS: Experimental ACO recapitulated features of both asthma and COPD, with mixed pulmonary eosinophilic/neutrophilic inflammation, small airway collagen deposition, and increased airway hyperresponsiveness. Transcriptomic analysis identified common and distinct dysregulated gene clusters in airway and parenchyma samples in experimental asthma, COPD, and ACO. Upstream driver analysis revealed increased expression of the transcription factor Spi1. Pharmacological inhibition of SPI1 using DB2313, reduced airway remodeling and airway hyperresponsiveness in experimental ACO. CONCLUSIONS: A new experimental model of ACO featuring chronic dual exposures to house dust mite and cigarette smoke mimics key disease features observed in patients with ACO and revealed novel disease mechanisms, including upregulation of SPI1, that are amenable to therapy.


Asunto(s)
Asma , Eosinofilia , Enfermedad Pulmonar Obstructiva Crónica , Hipersensibilidad Respiratoria , Animales , Femenino , Ratones , ARN , Factores de Transcripción , Transcriptoma
3.
J Clin Invest ; 132(11)2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35642632

RESUMEN

Although aging and lung injury are linked to the development of idiopathic pulmonary fibrosis (IPF), the underlying pathognomonic processes predisposing to fibrotic lesions remain largely unknown. A deficiency in the ability of type 2 alveolar epithelial cell (AEC2) progenitors to regenerate and repair the epithelia has been proposed as a critical factor. In this issue of the JCI, Liang et al. identify a deficiency in the zinc transporter SLC39A8 (ZIP8) in AEC2s and in the subsequent activation of the sirtuin SIRT1 that predisposes to decreased AEC2 renewal capacity and enhanced lung fibrosis in both IPF and aging lungs. Interestingly, the authors demonstrate the efficacy of modulating dietary zinc levels, suggesting the need for clinical trials to evaluate the therapeutic potential of dietary supplementation and the development of pharmacological modulation of the Zn/ZIP8/SIRT1 axis for treatment.


Asunto(s)
Proteínas de Transporte de Catión , Fibrosis Pulmonar Idiopática , Sirtuina 1 , Células Epiteliales Alveolares/metabolismo , Proteínas Portadoras , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Sirtuina 1/genética , Sirtuina 1/metabolismo
5.
Respirology ; 26(11): 1049-1059, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34472161

RESUMEN

BACKGROUND AND OBJECTIVE: Influenza virus (FLU), rhinovirus (RV) and respiratory syncytial virus (RSV) are the most common acute respiratory infections worldwide. Infection can cause severe health outcomes, while therapeutic options are limited, primarily relieving symptoms without attenuating the development of lesions or impaired lung function. We therefore examined the inflammatory response to these infections with the intent to identify common components that are critical drivers of immunopathogenesis and thus represent potential therapeutic targets. METHODS: BALB/c mice were infected with FLU, RV or RSV, and lung function, airway inflammation and immunohistopathology were measured over a 10-day period. Anti-IL-17A mAb was administered to determine the impact of attenuating this cytokine's function on the development and severity of disease. RESULTS: All three viruses induced severe airway constriction and inflammation at 2 days post-infection (dpi). However, only FLU induced prolonged inflammation till 10 dpi. Increased IL-17A expression was correlated with the alterations in lung function and its persistence. Neutralization of IL-17A did not affect the viral replication but led to the resolution of airway hyperresponsiveness. Furthermore, anti-IL-17A treatment resulted in reduced infiltration of neutrophils (in RV- and FLU-infected mice at 2 dpi) and lymphocytes (in RSV-infected mice at 2 dpi and FLU-infected mice at 10 dpi), and attenuated the severity of immunopathology. CONCLUSION: IL-17A is a common pathogenic molecule regulating disease induced by three prevalent respiratory viruses. Targeting the IL-17A pathway may provide a unified approach to the treatment of these respiratory infections alleviating both inflammation-induced lesions and difficulties in breathing.


Asunto(s)
Interleucina-17/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Picornaviridae/inmunología , Infecciones por Virus Sincitial Respiratorio , Animales , Pulmón/fisiopatología , Ratones , Ratones Endogámicos BALB C , Orthomyxoviridae , Virus Sincitiales Respiratorios/inmunología , Rhinovirus
6.
Mucosal Immunol ; 14(5): 1077-1087, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34083747

RESUMEN

CD4+ T-helper 22 (Th22) cells are a phenotypically distinct lymphocyte subset that produces high levels of interleukin (IL)-22 without co-production of IL-17A. However, the developmental origin and lineage classification of Th22 cells, their interrelationship to Th17 cells, and potential for plasticity at sites of infection and inflammation remain largely undefined. An improved understanding of the mechanisms underpinning the outgrowth of Th22 cells will provide insights into their regulation during homeostasis, infection, and disease. To address this knowledge gap we generated 'IL-17A-fate-mapping IL-17A/IL-22 reporter transgenic mice' and show that Th22 cells develop in the gastrointestinal tract and lung during bacterial infection without transitioning via an Il17a-expressing intermediate, although in some compartments alternative transition pathways exist. Th22-cell development was not dependent on T-bet; however, this transcription factor functioned as a promiscuous T-cell-intrinsic regulator of IL-17A and IL-22 production, in addition to regulating the outgrowth, phenotypic stability, and plasticity of Th22 cells. Thus, we demonstrate that at sites of mucosal bacterial infection Th22 cells develop as a distinct lineage independently of Th17 cells; though both lineages exhibit bidirectional phenotypic flexibility within infected tissues and their draining lymph nodes, and that T-bet plays a critical regulatory role in Th22-cell function and identity.


Asunto(s)
Infecciones Bacterianas/etiología , Infecciones Bacterianas/metabolismo , Diferenciación Celular/inmunología , Interleucinas/biosíntesis , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/fisiología , Células Th17/citología , Células Th17/metabolismo , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Inmunofenotipificación , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Subgrupos de Linfocitos T/citología , Interleucina-22
7.
J Clin Invest ; 131(7)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33792564

RESUMEN

Allergic asthma is a chronic inflammatory airway disease characterized by dysregulated type 2 immune responses, including degranulating airway eosinophils that induce tissue damage and airway hyperresponsiveness (AHR). The type 2 cytokines interleukin 5 (IL-5) and IL-13 and the eosinophil-specific chemokine CCL11/CCL24/CCL26 axis recruit, activate, and regulate eosinophils in the airways. In this issue of the JCI, Karcz et al. identified a mechanism involving the nucleotide sugar UDP-glucose (UDP-G) and the purinergic receptor P2Y14R in amplifying eosinophil accumulation in the lung. During type 2 inflammation, UDP-G activates P2Y14R on eosinophils, inducing the cells to move and migrate into the lung. Pharmacologically or genetically inhibiting P2Y14R on eosinophils attenuated eosinophil infiltration and AHR. Future experiments, including identifying additional type 2 factors regulating P2Y14R expression on lung eosinophils, are necessary to ascertain the impact of targeting P2Y14R as an alternative or adjunctive therapy to current type 2 biologics for the treatment of asthma.


Asunto(s)
Asma , Eosinófilos , Asma/tratamiento farmacológico , Asma/genética , Glucosa , Humanos , Interleucina-13 , Uridina Difosfato Glucosa
9.
Chest ; 158(2): 828-829, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32768068
10.
FASEB J ; 34(6): 7718-7732, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32293760

RESUMEN

Liver inflammation is a common extraintestinal manifestation in inflammatory bowel disease (IBD), yet, the mechanisms driving gut-liver axis inflammation remain poorly understood. IBD leads to a breakdown in the integrity of the intestinal barrier causing an increase in portal and systemic gut-derived antigens, which challenge the liver. Here, we examined the role of platelet activating factor receptor (PAFR) in colitis-associated liver damage using dextran sulfate sodium (DSS) and anti-CD40-induced colitis models. Both DSS and anti-CD40 models exhibited liver inflammation associated with colitis. Colitis reduced global PAFR protein expression in mouse livers causing an exclusive re-localization of PAFR to the portal triad. The global decrease in liver PAFR was associated with increased sirtuin 1 while relocalized PAFR expression was limited to Kupffer cells (KCs) and co-localized with toll-like receptor 4. DSS activated the NLRP3-inflammasome and increased interleukin (IL)-1ß in the liver. Antagonism of PAFR amplified the inflammasome response by increasing NLRP3, caspase-1, and IL-1ß protein levels in the liver. LPS also increased NLRP3 response in human hepatocytes, however, overexpression of PAFR restored the levels of NLPR3 and caspase-1 proteins. Interestingly, KCs depletion also increased IL-1ß protein in mouse liver after DSS challenge. These data suggest a protective role for PAFR-expressing KCs during colitis and that regulation of PAFR is important for gut-liver axis homeostasis.


Asunto(s)
Colitis/metabolismo , Colitis/patología , Inflamación/metabolismo , Inflamación/patología , Hígado/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Caspasa 1/metabolismo , Células Cultivadas , Colitis/inducido químicamente , Colon/metabolismo , Colon/patología , Sulfato de Dextran/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Interleucina-1beta/metabolismo , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor Toll-Like 4/metabolismo
11.
Pharmacol Res ; 158: 104782, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32275962

RESUMEN

Asthma is now recognised as a heterogenous inflammatory disease of the lung based on cellular infiltrates and transcriptional profiles of blood and airway cells. Four distinct subgroups have been defined, eosinophilic (T2), neutrophilic (T1), mixed eosinophilic/neutrophilic and paucigranulocytic. Patients can also be stratified at a molecular level into T2-high, T2-low and/or T1 based on their gene signatures. Current treatments for asthma have been centred on administration of steroids and/or bronchodilators for the relief of bronchoconstriction and inflammation. These treatments are not always effective and often have limited efficacy during exacerbations. Eosinophil expansion and homing to tissues, bronchoconstriction, IgE production and mucus hypersecretion (hallmark features of asthma) are regulated by the type 2 cytokines IL-4, IL-5 and IL-13, the latter of which can induce the expression of the eosinophil chemotactic factors CCL11 and CCL24. A number of new generation biologics (monoclonal antibodies) targeting pathways regulated by the T2 cytokines IL-5 and IL-4/13 (IL-4 receptor alpha) have yielded effective therapies for eosinophil induced exacerbations of severe asthma. Despite these advances, difficulties still remain in treating all exacerbations, and this may reflect the contribution of other inflammatory cells such as neutrophils to pathogenesis. This review describes the effectiveness of targeting T2 pathways, emerging approaches and identifies the potential next steps for therapeutic intervention.


Asunto(s)
Asma/terapia , Productos Biológicos/uso terapéutico , Inmunoterapia/métodos , Animales , Antiasmáticos/uso terapéutico , Asma/tratamiento farmacológico , Asma/fisiopatología , Citocinas/antagonistas & inhibidores , Eosinófilos/inmunología , Humanos , Transducción de Señal/efectos de los fármacos
12.
Eur Respir J ; 55(4)2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32184317

RESUMEN

Accumulating evidence highlights links between iron regulation and respiratory disease. Here, we assessed the relationship between iron levels and regulatory responses in clinical and experimental asthma.We show that cell-free iron levels are reduced in the bronchoalveolar lavage (BAL) supernatant of severe or mild-moderate asthma patients and correlate with lower forced expiratory volume in 1 s (FEV1). Conversely, iron-loaded cell numbers were increased in BAL in these patients and with lower FEV1/forced vital capacity (FVC) ratio. The airway tissue expression of the iron sequestration molecules divalent metal transporter 1 (DMT1) and transferrin receptor 1 (TFR1) are increased in asthma, with TFR1 expression correlating with reduced lung function and increased Type-2 (T2) inflammatory responses in the airways. Furthermore, pulmonary iron levels are increased in a house dust mite (HDM)-induced model of experimental asthma in association with augmented Tfr1 expression in airway tissue, similar to human disease. We show that macrophages are the predominant source of increased Tfr1 and Tfr1+ macrophages have increased Il13 expression. We also show that increased iron levels induce increased pro-inflammatory cytokine and/or extracellular matrix (ECM) responses in human airway smooth muscle (ASM) cells and fibroblasts ex vivo and induce key features of asthma in vivo, including airway hyper-responsiveness (AHR) and fibrosis, and T2 inflammatory responses.Together these complementary clinical and experimental data highlight the importance of altered pulmonary iron levels and regulation in asthma, and the need for a greater focus on the role and potential therapeutic targeting of iron in the pathogenesis and severity of disease.


Asunto(s)
Asma , Animales , Humanos , Interleucina-13 , Hierro , Pulmón , Pyroglyphidae
13.
Clin Exp Allergy ; 50(1): 82-94, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31579973

RESUMEN

BACKGROUND: Acute exacerbations of asthma represent a major burden of disease and are often caused by respiratory infections. Viral infections are recognized as significant triggers of exacerbations; however, less is understood about the how microbial bioproducts such as the endotoxin (lipopolysaccharide (LPS)) trigger episodes. Indeed, increased levels of LPS have been linked to asthma onset, severity and steroid resistance. OBJECTIVE: The goal of this study was to identify mechanisms underlying bacterial-induced exacerbations by employing LPS as a surrogate for infection. METHODS: We developed a mouse model of LPS-induced exacerbation on the background of pre-existing type-2 allergic airway disease (AAD). RESULTS: LPS-induced exacerbation was characterized by steroid-resistant airway hyperresponsiveness (AHR) and an exaggerated inflammatory response distinguished by increased numbers of infiltrating neutrophils/macrophages and elevated production of lung inflammatory cytokines, including TNFα, IFNγ, IL-27 and MCP-1. Expression of the type-2 associated inflammatory factors such as IL-5 and IL-13 were elevated in AAD but not altered by LPS exposure. Furthermore, AHR and airway inflammation were no longer suppressed by corticosteroid (dexamethasone) treatment after LPS exposure. Depletion of pulmonary macrophages by administration of 2-chloroadenosine into the lungs suppressed AHR and reduced IL-13, TNFα and IFNγ expression. Blocking IL-13 function, through either IL-13-deficiency or administration of specific blocking antibodies, also suppressed AHR and airway inflammation. CONCLUSIONS & CLINICAL RELEVANCE: We present evidence that IL-13 and innate immune pathways (in particular pulmonary macrophages) contribute to LPS-induced exacerbation of pre-existing AAD and provide insight into the complex molecular processes potentially underlying microbial-induced exacerbations.


Asunto(s)
Asma/inmunología , Dexametasona/farmacología , Glucocorticoides/farmacología , Interleucina-13/inmunología , Lipopolisacáridos/farmacología , Activación de Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Hipersensibilidad Respiratoria/inmunología , Resistencia de las Vías Respiratorias/efectos de los fármacos , Animales , Infecciones Bacterianas , Líquido del Lavado Bronquioalveolar/citología , Quimiocina CCL2 , Citocinas/efectos de los fármacos , Citocinas/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Medicamentos , Interferón gamma/efectos de los fármacos , Interferón gamma/inmunología , Interleucinas/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Ratones , Mucina 5AC/efectos de los fármacos , Mucina 5AC/metabolismo , Ovalbúmina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
14.
Front Immunol ; 11: 598165, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33597946

RESUMEN

Background: The anti-inflammatory effect of an α7nAChR agonist, PNU-282987, has previously been explored in the context of inflammatory disease. However, the effects of PNU-282987 on type 2 innate lymphoid cells (ILC2s)-mediated allergic airway inflammation has not yet been established. Aims: To determine the effects of PNU-282987 on the function of ILC2s in the context of IL-33- or Alternaria Alternata (AA)- induced airway inflammation. Methods: PNU-282987 was administered to mice that received recombinant IL-33 or AA intranasal challenges. Lung histological analysis and flow cytometry were performed to determine airway inflammation and the infiltration and activation of ILC2s. The previously published α7nAChR agonist GTS-21 was employed as a comparable reagent. ILC2s were isolated from murine lung tissue and cultured in vitro in the presence of IL-33, IL-2, and IL-7 with/without either PNU-282987 or GTS-21. The expression of the transcription factors GATA3, IKK, and NF-κB were also determined. Results: PNU-282987 and GTS-21 significantly reduced goblet cell hyperplasia in the airway, eosinophil infiltration, and ILC2s numbers in BALF, following IL-33 or AA challenge. In vitro IL-33 stimulation of isolated lung ILC2s showed a reduction of GATA3 and Ki67 in response to PNU-282987 or GTS-21 treatments. There was a significant reduction in IKK and NF-κB phosphorylation in the PNU-282987-treated group when compared to the GTS-21-treated ILC2s. Conclusion: PNU-282987 inhibits ILC2-associated airway inflammation, where its effects were comparable to that of GTS-21.


Asunto(s)
Asma/etiología , Asma/metabolismo , Benzamidas/farmacología , Compuestos Bicíclicos con Puentes/farmacología , Inmunidad Innata/efectos de los fármacos , Linfocitos/efectos de los fármacos , Linfocitos/fisiología , Agonistas Nicotínicos/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Animales , Asma/tratamiento farmacológico , Asma/patología , Compuestos de Bencilideno/farmacología , Biomarcadores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Inmunofenotipificación , Interleucina-33/metabolismo , Ratones , Piridinas/farmacología
15.
Chest ; 156(5): 991-1000, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31255581

RESUMEN

Chronic inflammatory diseases of the lung are often life-threatening and are a leading cause of morbidity in our communities. MicroRNAs (miRs) are now recognized to play critical roles in a wide range of cellular functions, including the regulation of immunologic processes, which are often dysregulated in chronic respiratory diseases. These small noncoding RNA molecules regulate networks of genes by inhibiting translation through the targeting of one or multiple messenger RNA transcripts. This review highlights discoveries that identify important roles for miRs in the regulation of specific pathogenic features of a range of diseases. Furthermore, experimental evidence suggests that pharmacologic inhibition of miR function or delivery of mimics may have therapeutic potential. The review also therefore discusses the potential utility and limitations of therapeutically targeting these molecules and their downstream pathways.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedades Pulmonares/genética , Pulmón/metabolismo , MicroARNs/genética , Animales , Progresión de la Enfermedad , Humanos , Enfermedades Pulmonares/metabolismo , MicroARNs/metabolismo , Transducción de Señal
16.
Mucosal Immunol ; 12(4): 862-873, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30976089

RESUMEN

Extra-intestinal manifestations (EIM) are common in inflammatory bowel disease (IBD). One such EIM is sub-clinical pulmonary inflammation, which occurs in up to 50% of IBD patients. In animal models of colitis, pulmonary inflammation is driven by neutrophilic infiltrations, primarily in response to the systemic bacteraemia and increased bacterial load in the lungs. Platelet activating factor receptor (PAFR) plays a critical role in regulating pulmonary responses to infection in conditions, such as chronic obstructive pulmonary disease and asthma. We investigated the role of PAFR in pulmonary EIMs of IBD, using dextran sulfate sodium (DSS) and anti-CD40 murine models of colitis. Both models induced neutrophilic inflammation, with increased TNF and IL-1ß levels, bacterial load and PAFR protein expression in mouse lungs. Antagonism of PAFR decreased lung neutrophilia, TNF, and IL-1ß in an NLRP3 inflammasome-dependent manner. Lipopolysaccharide from phosphorylcholine (ChoP)-positive bacteria induced NLRP3 and caspase-1 proteins in human alveolar epithelial cells, however antagonism of PAFR prevented NLRP3 activation by ChoP. Amoxicillin reduced bacterial populations in the lungs and reduced NLRP3 inflammasome protein levels, but did not reduce PAFR. These data suggest a role for PAFR in microbial pattern recognition and NLRP3 inflammasome signaling in the lung.


Asunto(s)
Colitis/complicaciones , Susceptibilidad a Enfermedades , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Neumonía/etiología , Neumonía/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Biopsia , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Endoscopía , Inmunohistoquímica , Inflamasomas/metabolismo , Enfermedades Inflamatorias del Intestino/complicaciones , Ratones , Infiltración Neutrófila/inmunología , Neumonía/patología , Transducción de Señal
17.
Am J Pathol ; 188(7): 1625-1639, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29684360

RESUMEN

Inflammatory bowel disease (IBD) is associated with several immune-mediated extraintestinal manifestations. More than half of all IBD patients have some form of respiratory pathology, most commonly neutrophil-mediated diseases, such as bronchiectasis and chronic bronchitis. Using murine models of colitis, we aimed to identify the immune mechanisms driving pulmonary manifestations of IBD. We found increased neutrophil numbers in lung tissue associated with the pulmonary vasculature in both trinitrobenzenesulfonic acid- and dextran sulfate sodium-induced models of colitis. Analysis of systemic inflammation identified that neutrophilia was associated with bacteremia and pyrexia in animal models of colitis. We further identified IL-6 as a systemic mediator of neutrophil recruitment from the bone marrow of dextran sulfate sodium animals. Functional inhibition of IL-6 led to reduced systemic and pulmonary neutrophilia, but it did not attenuate established colitis pathology. These data suggest that systemic bacteremia and pyrexia drive IL-6 secretion, which is a critical driver for pulmonary manifestation of IBD. Targeting IL-6 may reduce neutrophil-associated extraintestinal manifestations in IBD patients.


Asunto(s)
Bacteriemia/patología , Colitis/complicaciones , Modelos Animales de Enfermedad , Interleucina-6/toxicidad , Neutrófilos/inmunología , Neumonía/patología , Animales , Bacteriemia/etiología , Bacteriemia/metabolismo , Colitis/inducido químicamente , Sulfato de Dextran/toxicidad , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Neumonía/etiología , Neumonía/metabolismo
18.
Front Immunol ; 9: 210, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29472933

RESUMEN

The Parkinson's disease-associated protein, Leucine-rich repeat kinase 2 (LRRK2), a known negative regulator of nuclear factor of activated T cells (NFAT), is expressed in myeloid cells such as macrophages and dendritic cells (DCs) and is involved in the host immune response against pathogens. Since, the Ca2+/NFAT/IL-2 axis has been previously found to regulate DC response to the fungus Aspergillus, we have investigated the role played by the kinase LRRK2 during fungal infection. Mechanistically, we found that in the early stages of the non-canonical autophagic response of DCs to the germinated spores of Aspergillus, LRRK2 undergoes progressive degradation and regulates NFAT translocation from the cytoplasm to the nucleus. Our results shed new light on the complexity of the Ca2+/NFAT/IL-2 pathway, where LRRK2 plays a role in controlling the immune response of DCs to Aspergillus.


Asunto(s)
Aspergilosis/inmunología , Aspergillus/inmunología , Autofagia/inmunología , Células Dendríticas/inmunología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/inmunología , Transducción de Señal/inmunología , Animales , Aspergilosis/microbiología , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Células Cultivadas , Células Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Interacciones Huésped-Parásitos/inmunología , Humanos , Interleucina-2/metabolismo , Microscopía Intravital , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Factores de Transcripción NFATC/metabolismo , Proteolisis , ARN Largo no Codificante/genética , ARN Largo no Codificante/inmunología , ARN Largo no Codificante/metabolismo , ARN Interferente Pequeño/metabolismo , Esporas Fúngicas/inmunología , Imagen de Lapso de Tiempo
19.
J Immunol ; 200(2): 632-642, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29212906

RESUMEN

A link between inflammatory disease and bone loss is now recognized. However, limited data exist on the impact of virus infection on bone loss and regeneration. Bone loss results from an imbalance in remodeling, the physiological process whereby the skeleton undergoes continual cycles of formation and resorption. The specific molecular and cellular mechanisms linking virus-induced inflammation to bone loss remain unclear. In the current study, we provide evidence that infection of mice with either lymphocytic choriomeningitis virus (LCMV) or pneumonia virus of mice (PVM) resulted in rapid and substantial loss of osteoblasts from the bone surface. Osteoblast ablation was associated with elevated levels of circulating inflammatory cytokines, including TNF-α, IFN-γ, IL-6, and CCL2. Both LCMV and PVM infections resulted in reduced osteoblast-specific gene expression in bone, loss of osteoblasts, and reduced serum markers of bone formation, including osteocalcin and procollagen type 1 N propeptide. Infection of Rag-1-deficient mice (which lack adaptive immune cells) or specific depletion of CD8+ T lymphocytes limited osteoblast loss associated with LCMV infection. By contrast, CD8+ T cell depletion had no apparent impact on osteoblast ablation in association with PVM infection. In summary, our data demonstrate dramatic loss of osteoblasts in response to virus infection and associated systemic inflammation. Further, the inflammatory mechanisms mediating viral infection-induced bone loss depend on the specific inflammatory condition.


Asunto(s)
Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Neumonía Murina/inmunología , Osteoblastos/virología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/virología , Animales , Biomarcadores , Médula Ósea/patología , Huesos/metabolismo , Huesos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Proteínas de Homeodominio/genética , Depleción Linfocítica , Ratones , Ratones Noqueados , Osteoblastos/inmunología , Osteogénesis
20.
J Immunol ; 200(1): 237-247, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29167232

RESUMEN

Respiratory syncytial virus (RSV) infection induces asthma exacerbations, which leads to worsening of clinical symptoms and may result in a sustained decline in lung function. Exacerbations are the main cause of morbidity and mortality associated with asthma, and significantly contribute to asthma-associated healthcare costs. Although glucocorticoids are used to manage exacerbations, some patients respond to them poorly. The underlying mechanisms associated with steroid-resistant exacerbations remain largely unknown. We have previously established a mouse model of RSV-induced exacerbation of allergic airways disease, which mimics hallmark clinical features of asthma. In this study, we have identified key roles for macrophage IFN-γ and IL-27 in the regulation of RSV-induced exacerbation of allergic airways disease. Production of IFN-γ and IL-27 was steroid-resistant, and neutralization of IFN-γ or IL-27 significantly suppressed RSV-induced steroid-resistant airway hyperresponsiveness and airway inflammation. We have previously implicated activation of pulmonary macrophage by TNF-α and/or MCP-1 in the mechanisms of RSV-induced exacerbation. Stimulation of pulmonary macrophages with TNF-α and/or MCP-1 induced expression of both IFN-γ and IL-27. Our findings highlight critical roles for IFN-γ and IL-27, downstream of TNF-α and MCP-1, in the mechanism of RSV-induced exacerbation. Thus, targeting the pathways that these factors activate may be a potential therapeutic approach for virus-induced asthma exacerbations.


Asunto(s)
Asma/inmunología , Interferón gamma/metabolismo , Interleucina-27/metabolismo , Macrófagos Alveolares/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Asma/complicaciones , Células Cultivadas , Quimiocina CCL2/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Activación de Macrófagos , Macrófagos Alveolares/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/complicaciones , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA