Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Thorax ; 76(1): 92-99, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33077618

RESUMEN

The lungs are exposed to a range of environmental toxins (including cigarette smoke, air pollution, asbestos) and pathogens (bacterial, viral and fungal), and most respiratory diseases are associated with local or systemic hypoxia. All of these adverse factors can trigger endoplasmic reticulum (ER) stress. The ER is a key intracellular site for synthesis of secretory and membrane proteins, regulating their folding, assembly into complexes, transport and degradation. Accumulation of misfolded proteins within the lumen results in ER stress, which activates the unfolded protein response (UPR). Effectors of the UPR temporarily reduce protein synthesis, while enhancing degradation of misfolded proteins and increasing the folding capacity of the ER. If successful, homeostasis is restored and protein synthesis resumes, but if ER stress persists, cell death pathways are activated. ER stress and the resulting UPR occur in a range of pulmonary insults and the outcome plays an important role in many respiratory diseases. The UPR is triggered in the airway of patients with several respiratory diseases and in corresponding experimental models. ER stress has been implicated in the initiation and progression of pulmonary fibrosis, and evidence is accumulating suggesting that ER stress occurs in obstructive lung diseases (particularly in asthma), in pulmonary infections (some viral infections and in the setting of the cystic fibrosis airway) and in lung cancer. While a number of small molecule inhibitors have been used to interrogate the role of the UPR in disease models, many of these tools have complex and off-target effects, hence additional evidence (eg, from genetic manipulation) may be required to support conclusions based on the impact of such pharmacological agents. Aberrant activation of the UPR may be linked to disease pathogenesis and progression, but at present, our understanding of the context-specific and disease-specific mechanisms linking these processes is incomplete. Despite this, the ability of the UPR to defend against ER stress and influence a range of respiratory diseases is becoming increasingly evident, and the UPR is therefore attracting attention as a prospective target for therapeutic intervention strategies.


Asunto(s)
Estrés del Retículo Endoplásmico , Enfermedades Pulmonares/metabolismo , Proteínas de la Membrana/fisiología , Humanos , Transducción de Señal
2.
J Virol ; 93(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30333178

RESUMEN

Rhinoviral infection is a common trigger of the excessive inflammation observed during exacerbations of asthma and chronic obstructive pulmonary disease. Rhinovirus (RV) recognition by pattern recognition receptors activates the mitogen-activated protein kinase (MAPK) pathways, which are common inducers of inflammatory gene production. A family of dual-specificity phosphatases (DUSPs) can regulate MAPK function, but their roles in rhinoviral infection are not known. We hypothesized that DUSPs would negatively regulate the inflammatory response to RV infection. Our results revealed that the p38 and c-Jun N-terminal kinase (JNK) MAPKs play key roles in the inflammatory response of epithelial cells to RV infection. Three DUSPs previously shown to have roles in innate immunity (DUSPs 1, 4, and 10) were expressed in primary bronchial epithelial cells, and one of them, DUSP10, was downregulated by RV infection. Small interfering RNA-mediated knockdown of DUSP10 identified a role for the protein in negatively regulating inflammatory cytokine production in response to interleukin-1ß (IL-1ß), alone and in combination with RV, without any effect on RV replication. This study identifies DUSP10 as an important regulator of airway inflammation in respiratory viral infection.IMPORTANCE Rhinoviruses are one of the causes of the common cold. In patients with asthma or chronic obstructive pulmonary disease, viral infections, including those with rhinovirus, are the commonest cause of exacerbations. Novel therapeutics to limit viral inflammation are clearly required. The work presented here identifies DUSP10 as an important protein involved in limiting the inflammatory response in the airway without affecting immune control of the virus.


Asunto(s)
Bronquios/virología , Fosfatasas de Especificidad Dual/metabolismo , Interleucina-1beta/farmacología , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Rhinovirus/patogenicidad , Bronquios/citología , Bronquios/inmunología , Células Cultivadas , Regulación hacia Abajo , Fosfatasas de Especificidad Dual/genética , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/virología , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Rhinovirus/inmunología
3.
Biochem Soc Trans ; 46(2): 361-369, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29523773

RESUMEN

Defining features of chronic airway diseases include abnormal and persistent inflammatory processes, impaired airway epithelial integrity and function, and increased susceptibility to recurrent respiratory infections. Phosphoinositide 3-kinases (PI3K) are lipid kinases, which contribute to multiple physiological and pathological processes within the airway, with abnormal PI3K signalling contributing to the pathogenesis of several respiratory diseases. Consequently, the potential benefit of targeting PI3K isoforms has received considerable attention, being viewed as a viable therapeutic option in inflammatory and infectious lung disorders. The class I PI3K isoform, PI3Kδ (Phosphoinositide 3-kinases δ) is of particular interest given its multiple roles in modulating innate and adaptive immune cell functions, airway inflammation and corticosteroid sensitivity. In this mini-review, we explore the role of PI3Kδ in airway inflammation and infection, focusing on oxidative stress, ER stress, histone deacetylase 2 and neutrophil function. We also describe the importance of PI3Kδ in adaptive immune cell function, as highlighted by the recently described Activated PI3K Delta Syndrome, and draw attention to some of the potential clinical applications and benefits of targeting this molecule.


Asunto(s)
Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedades Respiratorias/enzimología , Animales , Linfocitos B/inmunología , Estrés del Retículo Endoplásmico , Histona Desacetilasa 2/metabolismo , Humanos , Inmunidad Innata , Inflamación/enzimología , Inflamación/inmunología , Células Mieloides/inmunología , Neutrófilos/inmunología , Estrés Oxidativo , Enfermedades Respiratorias/inmunología , Transducción de Señal , Linfocitos T/inmunología
4.
PLoS One ; 9(12): e116055, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25541728

RESUMEN

Human rhinoviruses (HRV) are a major cause of exacerbations of airways disease. Aspects of cell signalling responses to HRV infection remain unclear, particularly with regard to signalling via PI3K, and the PI3K-dependent pathway, autophagy. We investigated the roles of PI3K and autophagy in the responses of epithelial cells to major and minor group HRV infection. The PI3K inhibitor 3-MA, commonly used to inhibit autophagy, markedly reduced HRV-induced cytokine induction. Further investigation of potential targets of 3-MA and comparison of results using this inhibitor to a panel of general and class I-selective PI3K inhibitors showed that several PI3Ks cooperatively regulate responses to HRV. Targeting by siRNA of the autophagy proteins Beclin-1, Atg7, LC3, alone or in combination, or targeting of the autophagy-specific class III PI3K had at most only modest effects on HRV-induced cell signalling as judged by induction of proinflammatory cytokine production. Our data indicate that PI3K and mTOR are involved in induction of proinflammatory cytokines after HRV infection, and that autophagy has little role in the cytokine response to HRV or control of HRV replication.


Asunto(s)
Autofagia , Células Epiteliales/microbiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Infecciones por Picornaviridae/enzimología , Infecciones por Picornaviridae/fisiopatología , Inhibidores de Proteínas Quinasas/farmacología , Rhinovirus/fisiología , Línea Celular , Citocinas/inmunología , Células Epiteliales/inmunología , Células Epiteliales/patología , Interacciones Huésped-Patógeno , Humanos , Fosfatidilinositol 3-Quinasas/inmunología , Infecciones por Picornaviridae/inmunología , Transducción de Señal , Serina-Treonina Quinasas TOR/inmunología
5.
J Leukoc Biol ; 93(1): 7-19, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22904343

RESUMEN

This study examined the establishment of neutrophilic inflammation in humans. We tested the hypotheses that neutrophil recruitment was associated with local CXCL8 production and that neutrophils themselves might contribute to the regulation of the size of the inflammatory response. Humans were challenged i.d. with endotoxin. Biopsies of these sites were examined for cytokine production and leukocyte recruitment by qPCR and IHC. Additional in vitro models of inflammation examined the ability of neutrophils to produce and sequester cytokines relevant to neutrophilic inflammation. i.d. challenge with 15 ng of a TLR4-selective endotoxin caused a local inflammatory response, in which 1% of the total biopsy area stained positive for neutrophils at 6 h, correlating with 100-fold up-regulation in local CXCL8 mRNA generation. Neutrophils themselves were the major source of the early cytokine IL-1ß. In vitro, neutrophils mediated CXCL8 but not IL-1ß clearance (>90% clearance of ≤2 nM CXCL8 over 24 h). CXCL8 clearance was at least partially receptor-dependent and modified by inflammatory context, preserved in models of viral infection but reduced in models of bacterial infection. In conclusion, in a human inflammatory model, neutrophils are rapidly recruited and may regulate the size and outcome of the inflammatory response through the uptake and release of cytokines and chemokines in patterns dependent on the underlying inflammatory stimulus.


Asunto(s)
Quimiocinas/metabolismo , Inflamación/metabolismo , Interleucina-1/metabolismo , Infiltración Neutrófila/inmunología , Neutrófilos/metabolismo , Animales , Western Blotting , Quimiocinas/inmunología , Endotoxinas/toxicidad , Humanos , Inmunohistoquímica , Inflamación/inducido químicamente , Inflamación/inmunología , Interleucina-1/inmunología , Interleucina-8/inmunología , Interleucina-8/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología
6.
J Virol ; 86(12): 6595-604, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22514342

RESUMEN

Pellino-1 has recently been identified as a regulator of interleukin-1 (IL-1) signaling, but its roles in regulation of responses of human cells to human pathogens are unknown. We investigated the potential roles of Pellino-1 in the airways. We show for the first time that Pellino-1 regulates responses to a human pathogen, rhinovirus minor group serotype 1B (RV-1B). Knockdown of Pellino-1 by small interfering RNA (siRNA) was associated with impaired production of innate immune cytokines such as CXCL8 from human primary bronchial epithelial cells in response to RV-1B, without impairment in production of antiviral interferons (IFN), and without loss of control of viral replication. Pellino-1 actions were likely to be independent of interleukin-1 receptor-associated kinase-1 (IRAK-1) regulation, since Pellino-1 knockdown in primary epithelial cells did not alter responses to IL-1 but did inhibit responses to poly(I·C), a Toll-like receptor 3 (TLR3) activator that does not signal via IRAK-1 to engender a response. These data indicate that Pellino-1 represents a novel target that regulates responses of human airways to human viral pathogens, independently of IRAK signaling. Neutralization of Pellino-1 may therefore provide opportunities to inhibit potentially harmful neutrophilic inflammation of the airways induced by respiratory viruses, without loss of control of the underlying viral infection.


Asunto(s)
Células Epiteliales/inmunología , Proteínas Nucleares/inmunología , Infecciones por Picornaviridae/inmunología , Rhinovirus/fisiología , Ubiquitina-Proteína Ligasas/inmunología , Adolescente , Adulto , Anciano , Línea Celular , Células Cultivadas , Células Epiteliales/virología , Femenino , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/virología , Rhinovirus/genética , Rhinovirus/inmunología , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
7.
J Virol ; 85(15): 7912-21, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21593174

RESUMEN

Rhinoviral infection is an important trigger of acute inflammatory exacerbations in patients with underlying airway disease. We have previously established that interleukin-1ß (IL-1ß) is central in the communication between epithelial cells and monocytes during the initiation of inflammation. In this study we explored the roles of IL-1ß and its signaling pathways in the responses of airway cells to rhinovirus-1B (RV-1B) and further determined how responses to RV-1B were modified in a model of bacterial coinfection. Our results revealed that IL-1ß dramatically potentiated RV-1B-induced proinflammatory responses, and while monocytes did not directly amplify responses to RV-1B alone, they played an important role in the responses observed with our coinfection model. MyD88 is the essential signaling adapter for IL-1ß and most Toll-like receptors. To examine the role of MyD88 in more detail, we created stable MyD88 knockdown epithelial cells using short hairpin RNA (shRNA) targeted to MyD88. We determined that IL-1ß/MyD88 plays a role in regulating RV-1B replication and the inflammatory response to viral infection of airway cells. These results identify central roles for IL-1ß and its signaling pathways in the production of CXCL8, a potent neutrophil chemoattractant, in viral infection. Thus, IL-1ß is a viable target for controlling the neutrophilia that is often found in inflammatory airway disease and is exacerbated by viral infection of the airways.


Asunto(s)
Interleucina-1beta/fisiología , Factor 88 de Diferenciación Mieloide/metabolismo , Infecciones por Picornaviridae/metabolismo , Rhinovirus/aislamiento & purificación , Transducción de Señal , Western Blotting , Comunicación Celular , Línea Celular , Efecto Citopatogénico Viral , Ensayo de Inmunoadsorción Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Factor 88 de Diferenciación Mieloide/genética , Infecciones por Picornaviridae/virología , Reacción en Cadena de la Polimerasa , Rhinovirus/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...