Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 42(8): e110597, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-36912165

RESUMEN

The immunoproteasome is a specialized type of proteasome involved in MHC class I antigen presentation, antiviral adaptive immunity, autoimmunity, and is also part of a broader response to stress. Whether the immunoproteasome is regulated by DNA stress, however, is not known. We here demonstrate that mitochondrial DNA stress upregulates the immunoproteasome and MHC class I antigen presentation pathway via cGAS/STING/type I interferon signaling resulting in cell autonomous activation of CD8+ T cells. The cGAS/STING-induced adaptive immune response is also observed in response to genomic DNA and is conserved in epithelial and mesenchymal cells of mice and men. In patients with idiopathic pulmonary fibrosis, chronic activation of the cGAS/STING-induced adaptive immune response in aberrant lung epithelial cells concurs with CD8+ T-cell activation in diseased lungs. Genetic depletion of the immunoproteasome and specific immunoproteasome inhibitors counteract DNA stress induced cytotoxic CD8+ T-cell activation. Our data thus unravel cytoplasmic DNA sensing via the cGAS/STING pathway as an activator of the immunoproteasome and CD8+ T cells. This represents a novel potential pathomechanism for pulmonary fibrosis that opens new therapeutic perspectives.


Asunto(s)
Inmunidad Adaptativa , Linfocitos T CD8-positivos , ADN Mitocondrial , Antígenos de Histocompatibilidad Clase I/genética , Inmunidad Innata , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo
2.
Eur Respir J ; 62(2)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37385655

RESUMEN

BACKGROUND: Virus infections drive COPD exacerbations and progression. Antiviral immunity centres on the activation of virus-specific CD8+ T-cells by viral epitopes presented on major histocompatibility complex (MHC) class I molecules of infected cells. These epitopes are generated by the immunoproteasome, a specialised intracellular protein degradation machine, which is induced by antiviral cytokines in infected cells. METHODS: We analysed the effects of cigarette smoke on cytokine- and virus-mediated induction of the immunoproteasome in vitro, ex vivo and in vivo using RNA and Western blot analyses. CD8+ T-cell activation was determined in co-culture assays with cigarette smoke-exposed influenza A virus (IAV)-infected cells. Mass-spectrometry-based analysis of MHC class I-bound peptides uncovered the effects of cigarette smoke on inflammatory antigen presentation in lung cells. IAV-specific CD8+ T-cell numbers were determined in patients' peripheral blood using tetramer technology. RESULTS: Cigarette smoke impaired the induction of the immunoproteasome by cytokine signalling and viral infection in lung cells in vitro, ex vivo and in vivo. In addition, cigarette smoke altered the peptide repertoire of antigens presented on MHC class I molecules under inflammatory conditions. Importantly, MHC class I-mediated activation of IAV-specific CD8+ T-cells was dampened by cigarette smoke. COPD patients exhibited reduced numbers of circulating IAV-specific CD8+ T-cells compared to healthy controls and asthmatics. CONCLUSION: Our data indicate that cigarette smoke interferes with MHC class I antigen generation and presentation and thereby contributes to impaired activation of CD8+ T-cells upon virus infection. This adds important mechanistic insight on how cigarette smoke mediates increased susceptibility of smokers and COPD patients to viral infections.


Asunto(s)
Fumar Cigarrillos , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Linfocitos T CD8-positivos , Antivirales , Fumar Cigarrillos/efectos adversos , Antígenos de Histocompatibilidad Clase I/metabolismo , Citocinas , Epítopos , Inmunidad
3.
Eur Respir J ; 59(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34561290

RESUMEN

BACKGROUND: Immune cells contain a specialised type of proteasome, i.e. the immunoproteasome, which is required for intracellular protein degradation. Immunoproteasomes are key regulators of immune cell differentiation, inflammatory activation and autoimmunity. Immunoproteasome function in peripheral immune cells might be altered by smoking and in chronic obstructive pulmonary disease (COPD), thereby affecting immune cell responses. METHODS: We analysed the expression and activity of proteasome complexes in peripheral blood mononuclear cells (PBMCs) isolated from healthy male young smokers as well as from patients with severe COPD and compared them with matching controls. RESULTS: Proteasome expression was upregulated in COPD patients as assessed by quantitative reverse transcriptase-PCR and mass spectrometry-based proteomic analysis. Proteasome activity was quantified using activity-based probes and native gel analysis. We observed distinct activation of immunoproteasomes in the peripheral blood cells of young male smokers and severely ill COPD patients. Native gel analysis and linear regression modelling confirmed robust activation and elevated assembly of 20S proteasomes, which correlated significantly with reduced lung function parameters in COPD patients. The immunoproteasome was distinctly activated in COPD patients upon inflammatory cytokine stimulation of PBMCs in vitro. Inhibition of the immunoproteasome reduced pro-inflammatory cytokine expression in COPD-derived blood immune cells. CONCLUSIONS: Given the crucial role of chronic inflammatory signalling and the emerging involvement of autoimmune responses in COPD, therapeutic targeting of the immunoproteasome might represent a novel therapeutic concept for COPD.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Leucocitos Mononucleares/metabolismo , Masculino , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica , Fumadores
4.
FASEB J ; 33(11): 12392-12408, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31431059

RESUMEN

Insufficient autophagy has been reported in idiopathic pulmonary fibrosis (IPF) lungs. Specific roles of autophagy-related proteins in lung fibrosis development remain largely unknown. Here, we investigated the role of autophagy marker protein microtubule-associated protein 1 light chain 3ß (LC3B) in the development of lung fibrosis. LC3B-/- mice upon aging show smaller lamellar body profiles, increased cellularity, alveolar epithelial cell type II (AECII) apoptosis, surfactant alterations, and lysosomal and endoplasmic reticulum stress. Autophagosomal soluble N-ethylmaleimide-sensitive factor attachment protein receptor syntaxin 17 is increased in the AECII of aged LC3B-/- mice and patients with IPF. Proteasomal activity, however, remained unaltered in LC3B-/- mice. In vitro knockdown of LC3B sensitized mouse lung epithelial cells to bleomycin-induced apoptosis, but its overexpression was protective. In vivo, LC3B-/- mice displayed increased susceptibility to bleomycin-induced lung injury and fibrosis. We identified cathepsin A as a novel LC3B binding partner and its overexpression in vitro drives MLE12 cells to apoptosis. Additionally, cathepsin A is increased in the AECII of aged LC3B-/- mice and in the lungs of patients with IPF. Our study reveals that LC3B mediated autophagy plays essential roles in AECII by modulating the functions of proteins like cathepsin A and protects alveolar epithelial cells from apoptosis and subsequent lung injury and fibrosis.-Kesireddy, V. S., Chillappagari, S., Ahuja, S., Knudsen, L., Henneke, I., Graumann, J., Meiners, S., Ochs, M., Ruppert, C., Korfei, M., Seeger, W., Mahavadi, P. Susceptibility of microtubule-associated protein 1 light chain 3ß (MAP1LC3B/LC3B) knockout mice to lung injury and fibrosis.


Asunto(s)
Células Epiteliales Alveolares , Apoptosis/genética , Predisposición Genética a la Enfermedad , Proteínas Asociadas a Microtúbulos/deficiencia , Fibrosis Pulmonar , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Bleomicina/efectos adversos , Bleomicina/farmacología , Catepsina A/genética , Catepsina A/metabolismo , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo
5.
Adv Exp Med Biol ; 1233: 55-100, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32274753

RESUMEN

The proteasome is involved in the regulation of all cellular pathways and consequently plays a central role in the control of cellular homeostasis. Together with its regulators, it is at the frontline, both as an actor and as a target, in human health and when homeostasis is disturbed in disease. In this review, we aim to provide an overview of the many levels at which the functions of the proteasome and its regulators can be regulated to cope with cellular needs or are altered in pathological conditions.


Asunto(s)
Enfermedad , Salud , Complejo de la Endopetidasa Proteasomal , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo
6.
Proteomics ; 17(1-2)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27891773

RESUMEN

Cigarette smoke is the most relevant risk factor for the development of lung cancer and chronic obstructive pulmonary disease. Many of its more than 4500 chemicals are highly reactive, thereby altering protein structure and function. Here, we used subcellular fractionation coupled to label-free quantitative MS to globally assess alterations in the proteome of different compartments of lung epithelial cells upon exposure to cigarette smoke extract. Proteomic profiling of the human alveolar derived cell line A549 revealed the most pronounced changes within the cellular secretome with preferential downregulation of proteins involved in wound healing and extracellular matrix organization. In particular, secretion of secreted protein acidic and rich in cysteine, a matricellular protein that functions in tissue response to injury, was consistently diminished by cigarette smoke extract in various pulmonary epithelial cell lines and primary cells of human and mouse origin as well as in mouse ex vivo lung tissue cultures. Our study reveals a previously unrecognized acute response of lung epithelial cells to cigarette smoke that includes altered secretion of proteins involved in extracellular matrix organization and wound healing. This may contribute to sustained alterations in tissue remodeling as observed in lung cancer and chronic obstructive pulmonary disease.


Asunto(s)
Células Epiteliales/metabolismo , Pulmón/citología , Fumar/efectos adversos , Línea Celular , Células Epiteliales/efectos de los fármacos , Humanos , Proteómica/métodos , Cicatrización de Heridas/efectos de los fármacos
7.
J Biol Chem ; 291(25): 13147-59, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27129254

RESUMEN

The proteasome is an intracellular protease complex consisting of the 20S catalytic core and its associated regulators, including the 19S complex, PA28αß, PA28γ, PA200, and PI31. Inhibition of the proteasome induces autoregulatory de novo formation of 20S and 26S proteasome complexes. Formation of alternative proteasome complexes, however, has not been investigated so far. We here show that catalytic proteasome inhibition results in fast recruitment of PA28γ and PA200 to 20S and 26S proteasomes within 2-6 h. Rapid formation of alternative proteasome complexes did not involve transcriptional activation of PA28γ and PA200 but rather recruitment of preexisting activators to 20S and 26S proteasome complexes. Recruitment of proteasomal activators depended on the extent of active site inhibition of the proteasome with inhibition of ß5 active sites being sufficient for inducing recruitment. Moreover, specific inhibition of 26S proteasome activity via siRNA-mediated knockdown of the 19S subunit RPN6 induced recruitment of only PA200 to 20S proteasomes, whereas PA28γ was not mobilized. Here, formation of alternative PA200 complexes involved transcriptional activation of the activator. Alternative proteasome complexes persisted when cells had regained proteasome activity after pulse exposure to proteasome inhibitors. Knockdown of PA28γ sensitized cells to proteasome inhibitor-mediated growth arrest. Thus, formation of alternative proteasome complexes appears to be a formerly unrecognized but integral part of the cellular response to impaired proteasome function and altered proteostasis.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Autoantígenos/metabolismo , Bortezomib/farmacología , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Nucleares/metabolismo , Oligopéptidos/farmacología , Inhibidores de Proteasoma/farmacología , Multimerización de Proteína , Transcripción Genética
8.
Eur Respir J ; 49(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28179435

RESUMEN

Proteases were traditionally viewed as mere protein-degrading enzymes with a very restricted spectrum of substrates. A major expansion in protease research has uncovered a variety of novel substrates, and it is now evident that proteases are critical pleiotropic actors orchestrating pathophysiological processes. Recent findings evidenced that the net proteolytic activity also relies upon interconnections between different protease and protease inhibitor families in the protease web.In this review, we provide an overview of these novel concepts with a particular focus on pulmonary pathophysiology. We describe the emerging roles of several protease families including cysteine and serine proteases.The complexity of the protease web is exemplified in the light of multidimensional regulation of serine protease activity by matrix metalloproteases through cognate serine protease inhibitor processing. Finally, we will highlight how deregulated protease activity during pulmonary pathogenesis may be exploited for diagnosis/prognosis purposes, and utilised as a therapeutic tool using nanotechnologies.Considering proteases as part of an integrative biology perspective may pave the way for the development of new therapeutic targets to treat pulmonary diseases related to intrinsic protease deregulation.


Asunto(s)
Enfermedades Pulmonares/enzimología , Pulmón/enzimología , Metaloproteinasas de la Matriz/metabolismo , Animales , Humanos , Pulmón/inmunología , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/inmunología , Ratones , Inhibidores de Proteasas/uso terapéutico , Proteolisis/efectos de los fármacos
9.
Am J Respir Crit Care Med ; 193(11): 1230-41, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26756824

RESUMEN

RATIONALE: Patients with chronic obstructive pulmonary disease (COPD) and in particular smokers are more susceptible to respiratory infections contributing to acute exacerbations of disease. The immunoproteasome is a specialized type of proteasome destined to improve major histocompatibility complex (MHC) class I-mediated antigen presentation for the resolution of intracellular infections. OBJECTIVES: To characterize immunoproteasome function in COPD and its regulation by cigarette smoke. METHODS: Immunoproteasome expression and activity were determined in bronchoalveolar lavage (BAL) and lungs of human donors and patients with COPD or idiopathic pulmonary fibrosis (IPF), as well as in cigarette smoke-exposed mice. Smoke-mediated alterations of immunoproteasome activity and MHC I surface expression were analyzed in human blood-derived macrophages. Immunoproteasome-specific MHC I antigen presentation was evaluated in spleen and lung immune cells that had been smoke-exposed in vitro or in vivo. MEASUREMENTS AND MAIN RESULTS: Immunoproteasome and MHC I mRNA expression was reduced in BAL cells of patients with COPD and in isolated alveolar macrophages of patients with COPD or IPF. Exposure of immune cells to cigarette smoke extract in vitro reduced immunoproteasome activity and impaired immunoproteasome-specific MHC I antigen presentation. In vivo, acute cigarette smoke exposure dynamically regulated immunoproteasome function and MHC I antigen presentation in mouse BAL cells. End-stage COPD lungs showed markedly impaired immunoproteasome activities. CONCLUSIONS: We here show that the activity of the immunoproteasome is impaired by cigarette smoke resulting in reduced MHC I antigen presentation. Regulation of immunoproteasome function by cigarette smoke may thus alter adaptive immune responses and add to prolonged infections and exacerbations in COPD and IPF.


Asunto(s)
Inmunoproteínas/fisiología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Humo/efectos adversos , Fumar/fisiopatología , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Nicotiana
10.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L328-36, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27343191

RESUMEN

The proteasome system degrades more than 80% of intracellular proteins into small peptides. Accordingly, the proteasome is involved in many essential cellular functions, such as protein quality control, transcription, immune responses, cell signaling, and apoptosis. Moreover, degradation products are loaded onto major histocompatibility class I molecules to communicate the intracellular protein composition to the immune system. The standard 20S proteasome core complex contains three distinct catalytic active sites that are exchanged upon stimulation with inflammatory cytokines to form the so-called immunoproteasome. Immunoproteasomes are constitutively expressed in immune cells and have different proteolytic activities compared with standard proteasomes. They are rapidly induced in parenchymal cells upon intracellular pathogen infection and are crucial for priming effective CD8(+) T-cell-mediated immune responses against infected cells. Beyond shaping these adaptive immune reactions, immunoproteasomes also regulate the function of immune cells by degradation of inflammatory and immune mediators. Accordingly, they emerge as novel regulators of innate immune responses. The recently unraveled impairment of immunoproteasome function by environmental challenges and by genetic variations of immunoproteasome genes might represent a currently underestimated risk factor for the development and progression of lung diseases. In particular, immunoproteasome dysfunction will dampen resolution of infections, thereby promoting exacerbations, may foster autoimmunity in chronic lung diseases, and possibly contributes to immune evasion of tumor cells. Novel pharmacological tools, such as site-specific inhibitors of the immunoproteasome, as well as activity-based probes, however, hold promises as innovative therapeutic drugs for respiratory diseases and biomarker profiling, respectively.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Complejo de la Endopetidasa Proteasomal/fisiología , Animales , Autoinmunidad , Humanos , Pulmón/enzimología , Pulmón/patología , Enfermedades Pulmonares/enzimología , Enfermedades Pulmonares/inmunología
11.
Clin Sci (Lond) ; 130(4): 273-87, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26564208

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterized by chronic bronchitis, small airway remodelling and emphysema. Emphysema is the destruction of alveolar structures, leading to enlarged airspaces and reduced surface area impairing the ability for gaseous exchange. To further understand the pathological mechanisms underlying progressive emphysema, we used MS-based approaches to quantify the lung, bronchoalveolar lavage fluid (BALF) and serum metabolome during emphysema progression in the established murine porcine pancreatic elastase (PPE) model on days 28, 56 and 161, compared with PBS controls. Partial least squares (PLS) analysis revealed greater changes in the metabolome of lung followed by BALF rather than serum during emphysema progression. Furthermore, we demonstrate for the first time that emphysema progression is associated with a reduction in lung-specific L-carnitine, a metabolite critical for transporting long-chain fatty acids into the mitochondria for their subsequent ß-oxidation. In vitro, stimulation of the alveolar epithelial type II (ATII)-like LA4 cell line with L-carnitine diminished apoptosis induced by both PPE and H2O2. Moreover, PPE-treated mice demonstrated impaired lung function compared with PBS-treated controls (lung compliance; 0.067±0.008 ml/cmH20 compared with 0.035±0.005 ml/cmH20, P<0.0001), which improved following supplementation with L-carnitine (0.051±0.006, P<0.01) and was associated with a reduction in apoptosis. In summary, our results provide a new insight into the role of L-carnitine and, importantly, suggest therapeutic avenues for COPD.


Asunto(s)
Carnitina/metabolismo , Pulmón/metabolismo , Metaboloma , Metabolómica , Enfisema Pulmonar/metabolismo , Animales , Apoptosis , Biomarcadores/sangre , Líquido del Lavado Bronquioalveolar/química , Carnitina/sangre , Carnitina/farmacología , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Análisis de los Mínimos Cuadrados , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Rendimiento Pulmonar , Espectrometría de Masas , Metabolómica/métodos , Ratones Endogámicos C57BL , Elastasa Pancreática , Enfisema Pulmonar/sangre , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/patología , Enfisema Pulmonar/fisiopatología , Enfisema Pulmonar/prevención & control , Superóxidos/metabolismo , Factores de Tiempo
12.
Am J Respir Crit Care Med ; 192(9): 1089-101, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26207697

RESUMEN

RATIONALE: The ubiquitin-proteasome system is critical for maintenance of protein homeostasis by degrading polyubiquitinated proteins in a spatially and temporally controlled manner. Cell and protein homeostasis are altered upon pathological tissue remodeling. Dysregulation of the proteasome has been reported for several chronic diseases of the heart, brain, and lung. We hypothesized that proteasome function is altered upon fibrotic lung remodeling, thereby contributing to the pathogenesis of idiopathic pulmonary fibrosis (IPF). OBJECTIVES: To investigate proteasome function during myofibroblast differentiation. METHODS: We treated lung fibroblasts with transforming growth factor (TGF)-ß and examined proteasome composition and activity. For in vivo analysis, we used mouse models of lung fibrosis and fibrotic human lung tissue. MEASUREMENTS AND MAIN RESULTS: We demonstrate that induction of myofibroblast differentiation by TGF-ß involves activation of the 26S proteasome, which is critically dependent on the regulatory subunit Rpn6. Silencing of Rpn6 in primary human lung fibroblasts counteracted TGF-ß-induced myofibroblast differentiation. Activation of the 26S proteasome and increased expression of Rpn6 were detected during bleomycin-induced lung remodeling and fibrosis. Importantly, Rpn6 is overexpressed in myofibroblasts and basal cells of the bronchiolar epithelium in lungs of patients with IPF, which is accompanied by enhanced protein polyubiquitination. CONCLUSIONS: We identified Rpn6-dependent 26S proteasome activation as an essential feature of myofibroblast differentiation in vitro and in vivo, and our results suggest it has an important role in IPF pathogenesis.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/fisiopatología , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Western Blotting , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/metabolismo , Pulmón/fisiopatología , Masculino , Ratones , Persona de Mediana Edad , Miofibroblastos/metabolismo , Transducción de Señal
13.
Am J Respir Crit Care Med ; 191(3): 261-9, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25590812

RESUMEN

The aging of the population in the United States and throughout the developed world has increased morbidity and mortality attributable to lung disease, while the morbidity and mortality from other prevalent diseases has declined or remained stable. Recognizing the importance of aging in the development of lung disease, the American Thoracic Society (ATS) highlighted this topic as a core theme for the 2014 annual meeting. The relationship between aging and lung disease was discussed in several oral symposiums and poster sessions at the annual ATS meeting. In this article, we used the input gathered at the conference to develop a broad framework and perspective to stimulate basic, clinical, and translational research to understand how the aging process contributes to the onset and/or progression of lung diseases. A consistent theme that emerged from the conference was the need to apply novel, systems-based approaches to integrate a growing body of genomic, epigenomic, transcriptomic, and proteomic data and elucidate the relationship between biologic hallmarks of aging, altered lung function, and increased susceptibility to lung diseases in the older population. The challenge remains to causally link the molecular and cellular changes of aging with age-related changes in lung physiology and disease susceptibility. The purpose of this review is to stimulate further research to identify new strategies to prevent or treat age-related lung disease.


Asunto(s)
Envejecimiento , Enfermedades Pulmonares/epidemiología , Congresos como Asunto , Susceptibilidad a Enfermedades , Genoma Humano , Humanos , Fibrosis Pulmonar Idiopática/epidemiología , Incidencia , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/mortalidad , Fenotipo , Prevalencia , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Factores de Riesgo , Transcriptoma , Estados Unidos/epidemiología
14.
Eur Respir J ; 45(3): 807-27, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25657021

RESUMEN

Ageing is the main risk factor for major non-communicable chronic lung diseases, including chronic obstructive pulmonary disease, most forms of lung cancer and idiopathic pulmonary fibrosis. While the prevalence of these diseases continually increases with age, their respective incidence peaks at different times during the lifespan, suggesting specific effects of ageing on the onset and/or pathogenesis of chronic obstructive pulmonary disease, lung cancer and idiopathic pulmonary fibrosis. Recently, the nine hallmarks of ageing have been defined as cell-autonomous and non-autonomous pathways involved in ageing. Here, we review the available evidence for the involvement of each of these hallmarks in the pathogenesis of chronic obstructive pulmonary disease, lung cancer, or idiopathic pulmonary fibrosis. Importantly, we propose an additional hallmark, "dysregulation of the extracellular matrix", which we argue acts as a crucial modifier of cell-autonomous changes and functions, and as a key feature of the above-mentioned lung diseases.


Asunto(s)
Envejecimiento/fisiología , Senescencia Celular/fisiología , Fibrosis Pulmonar Idiopática , Neoplasias Pulmonares , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Pulmón/fisiopatología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Transducción de Señal/fisiología
15.
Eur Respir J ; 46(4): 1150-66, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25929950

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterised by a progressive loss of lung tissue. Inducing repair processes within the adult diseased lung is of major interest and Wnt/ß-catenin signalling represents a promising target for lung repair. However, the translation of novel therapeutic targets from model systems into clinical use remains a major challenge.We generated murine and patient-derived three-dimensional (3D) ex vivo lung tissue cultures (LTCs), which closely mimic the 3D lung microenvironment in vivo. Using two well-known glycogen synthase kinase-3ß inhibitors, lithium chloride (LiCl) and CHIR 99021 (CT), we determined Wnt/ß-catenin-driven lung repair processes in high spatiotemporal resolution using quantitative PCR, Western blotting, ELISA, (immuno)histological assessment, and four-dimensional confocal live tissue imaging.Viable 3D-LTCs exhibited preserved lung structure and function for up to 5 days. We demonstrate successful Wnt/ß-catenin signal activation in murine and patient-derived 3D-LTCs from COPD patients. Wnt/ß-catenin signalling led to increased alveolar epithelial cell marker expression, decreased matrix metalloproteinase-12 expression, as well as altered macrophage activity and elastin remodelling. Importantly, induction of surfactant protein C significantly correlated with disease stage (per cent predicted forced expiratory volume in 1 s) in patient-derived 3D-LTCs.Patient-derived 3D-LTCs represent a valuable tool to analyse potential targets and drugs for lung repair. Enhanced Wnt/ß-catenin signalling attenuated pathological features of patient-derived COPD 3D-LTCs.


Asunto(s)
Pulmón/citología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Proteínas Wnt/metabolismo , Adulto , Anciano , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Enfisema/fisiopatología , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/citología , Femenino , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Humanos , Cloruro de Litio/química , Pulmón/fisiopatología , Macrófagos Alveolares/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Piridinas/química , Pirimidinas/química , Transducción de Señal , Porcinos , Cicatrización de Heridas , beta Catenina/metabolismo
16.
Am J Respir Cell Mol Biol ; 50(6): 1040-52, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24358952

RESUMEN

The airway epithelium constitutes an essential immunological and cytoprotective barrier to inhaled insults, such as cigarette smoke, environmental particles, or viruses. Although bronchial epithelial integrity is crucial for airway homeostasis, defective epithelial barrier function contributes to chronic obstructive pulmonary disease (COPD). Tight junctions at the apical side of epithelial cell-cell contacts determine epithelial permeability. Cigarette smoke exposure, the major risk factor for COPD, is suggested to impair tight junction integrity; however, detailed mechanisms thereof remain elusive. We investigated whether cigarette smoke extract (CSE) and transforming growth factor (TGF)-ß1 affected tight junction integrity. Exposure of human bronchial epithelial cells (16HBE14o(-)) and differentiated primary human bronchial epithelial cells (pHBECs) to CSE significantly disrupted tight junction integrity and barrier function. Specifically, CSE decreased transepithelial electrical resistance (TEER) and tight junction-associated protein levels. Zonula occludens (ZO)-1 and ZO-2 protein levels were significantly reduced and dislocated from the cell membrane, as observed by fractionation and immunofluorescence analysis. These findings were reproduced in isolated bronchi exposed to CSE ex vivo, as detected by real-time quantitative reverse-transcriptase PCR and immunohistochemistry. Combined treatment of 16HBE14o(-) cells or pHBECs with CSE and TGF-ß1 restored ZO-1 and ZO-2 levels. TGF-ß1 cotreatment restored membrane localization of ZO-1 and ZO-2 protein and prevented CSE-mediated TEER decrease. In conclusion, CSE led to the disruption of tight junctions of human bronchial epithelial cells, and TGF-ß1 counteracted this CSE-induced effect. Thus, TGF-ß1 may serve as a protective factor for bronchial epithelial cell homeostasis in diseases such as COPD.


Asunto(s)
Bronquios/efectos de los fármacos , Nicotiana/efectos adversos , Humo/efectos adversos , Uniones Estrechas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Animales , Bronquios/metabolismo , Diferenciación Celular/genética , Línea Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Mucosa Respiratoria/metabolismo , Fumar/efectos adversos , Uniones Estrechas/genética , Factor de Crecimiento Transformador beta1/genética , Regulación hacia Arriba , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo , Proteína de la Zonula Occludens-2/genética , Proteína de la Zonula Occludens-2/metabolismo
17.
Am J Respir Cell Mol Biol ; 51(4): 526-35, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24773184

RESUMEN

In inhalation therapy, drugs are deposited as aerosols onto the air-facing lung epithelium. The currently used in vitro cell assays for drug testing, however, typically dissolve drugs in the medium, completely covering the cells, which represents an unphysiological drug application scenario. Although physiologically realistic in vitro cell culture models of the pulmonary air-blood barrier are available, reliable, easy-to-handle, and efficient technologies for direct aerosol-to-cell delivery are lacking. Here, we introduce the Air-Liquid Interface (ALI) Cell Exposure-Cloud (ALICE-CLOUD) technology, which uses principles of cloud motion for fast and quantitative delivery of aerosolized liquid drugs to pulmonary cells cultured under realistic ALI conditions. Aerosol-to-cell delivery proved to be highly efficient, reproducible, and rapid when using aerosolized fluorescein as surrogate drug. As a proof-of-concept study for the ALICE-CLOUD, we performed functional efficacy studies with the U.S. Food and Drug Administration-approved proteasome inhibitor, Bortezomib, a novel candidate drug for inhalation therapy. Aerosolized Bortezomib had a pronounced anti-inflammatory effect on human epithelial lung cells (A549), as indicated by a significant reduction of (TNFα-induced) IL-8 promoter activation. Importantly, cell-based therapeutic efficacy of aerosolized Bortezomib under ALI conditions was similar to that under dissolved and nonaerosolized submerged conditions, but with faster uptake kinetics. Our data indicate that the ALICE-CLOUD is a reliable tool for aerosolized drug screening with cells cultured under ALI conditions, which combines ease of handling with rapid, efficient, and dosimetrically accurate drug-to-cell delivery. This may pave the way for screening of inhalable drugs under physiologically more relevant and, hence, potentially more predictive conditions than the currently used submerged cell culture systems.


Asunto(s)
Antiinflamatorios/administración & dosificación , Barrera Alveolocapilar/efectos de los fármacos , Ácidos Borónicos/administración & dosificación , Células Epiteliales/efectos de los fármacos , Inhibidores de Proteasoma/administración & dosificación , Pirazinas/administración & dosificación , Mucosa Respiratoria/efectos de los fármacos , Administración por Inhalación , Aerosoles , Antiinflamatorios/metabolismo , Barrera Alveolocapilar/inmunología , Barrera Alveolocapilar/metabolismo , Ácidos Borónicos/metabolismo , Bortezomib , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Cinética , Regiones Promotoras Genéticas , Inhibidores de Proteasoma/metabolismo , Pirazinas/metabolismo , Reproducibilidad de los Resultados , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
18.
Am J Physiol Lung Cell Mol Physiol ; 307(11): L895-907, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25326581

RESUMEN

Cigarette smoke is the main risk factor for chronic obstructive pulmonary disease (COPD). Exposure of cells to cigarette smoke induces an initial adaptive cellular stress response involving increased oxidative stress and induction of inflammatory signaling pathways. Exposure of mitochondria to cellular stress alters their fusion/fission dynamics. Whereas mild stress induces a prosurvival response termed stress-induced mitochondrial hyperfusion, severe stress results in mitochondrial fragmentation and mitophagy. In the present study, we analyzed the mitochondrial response to mild and nontoxic doses of cigarette smoke extract (CSE) in alveolar epithelial cells. We characterized mitochondrial morphology, expression of mitochondrial fusion and fission genes, markers of mitochondrial proteostasis, as well as mitochondrial functions such as membrane potential and oxygen consumption. Murine lung epithelial (MLE)12 and primary mouse alveolar epithelial cells revealed pronounced mitochondrial hyperfusion upon treatment with CSE, accompanied by increased expression of the mitochondrial fusion protein mitofusin 2 and increased metabolic activity. We did not observe any alterations in mitochondrial proteostasis, i.e., induction of the mitochondrial unfolded protein response or mitophagy. Therefore, our data indicate an adaptive prosurvival response of mitochondria of alveolar epithelial cells to nontoxic concentrations of CSE. A hyperfused mitochondrial network, however, renders the cell more vulnerable to additional stress, such as sustained cigarette smoke exposure. As such, cigarette smoke-induced mitochondrial hyperfusion, although part of a beneficial adaptive stress response in the first place, may contribute to the pathogenesis of COPD.


Asunto(s)
Mitocondrias/efectos de los fármacos , Nicotiana/efectos adversos , Alveolos Pulmonares/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/etiología , Mucosa Respiratoria/efectos de los fármacos , Humo/efectos adversos , Animales , Línea Celular , GTP Fosfohidrolasas/biosíntesis , Inflamación/inducido químicamente , Pulmón/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mitofagia/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Alveolos Pulmonares/ultraestructura , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/ultraestructura , Fumar/efectos adversos , Respuesta de Proteína Desplegada/efectos de los fármacos
19.
Eur Respir J ; 44(3): 765-74, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24791828

RESUMEN

Respiratory diseases are an increasing burden for the ageing population. Although our understanding of these diseases has improved significantly over the past decades, diagnostic and therapeutic options for treating lung diseases, such as chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis and lung cancer, remain limited. Multidisciplinary approaches that bridge the gap between medicinal and materials sciences will likely contribute to promising new therapeutic and diagnostic solutions. One such multidisciplinary approach is the use of nanoparticles as carriers for the delivery of drugs. The advantages of using nanoparticles to deliver drugs include: increased drug concentration at the disease site; minimised drug degradation and loss; ease of creating inhalable formulations; and the possibility of specific cell targeting. This article gives a brief overview on the emerging field of nanocarriers as drug delivery vehicles for the treatment of lung diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos , Enfermedades Pulmonares/tratamiento farmacológico , Pulmón/efectos de los fármacos , Nanomedicina/métodos , Nanopartículas/química , Química Farmacéutica/tendencias , Portadores de Fármacos , Humanos , Pulmón/fisiopatología , Neoplasias Pulmonares/tratamiento farmacológico , Preparaciones Farmacéuticas/química , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Tecnología Farmacéutica/tendencias
20.
Breathe (Sheff) ; 20(1): 230183, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38746906

RESUMEN

The Translational Science Working Group at the European Respiratory Society (ERS) aims to bridge the gap between basic and clinical science by providing a platform where scientists, clinicians and experts in the respiratory field can actively shape translational research. For the 2023 Congress, dedicated translational science sessions were created and sessions of interest to many assemblies from the clinical and the scientific point of view were tagged as translational sessions, attracting clinical and scientific experts to the same room to discuss relevant topics and strengthening translational efforts among all ERS assemblies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA