Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ERJ Open Res ; 10(1)2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38375425

RESUMEN

Introduction: Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial pneumonia marked by progressive lung fibrosis and a poor prognosis. Recent studies have highlighted the potential role of infection in the pathogenesis of IPF, and a prior association of the HLA-DQB1 gene with idiopathic fibrotic interstitial pneumonia (including IPF) has been reported. Owing to the important role that the human leukocyte antigen (HLA) region plays in the immune response, here we evaluated if HLA genetic variation was associated specifically with IPF risk. Methods: We performed a meta-analysis of associations of the HLA region with IPF risk in individuals of European ancestry from seven independent case-control studies of IPF (comprising 5159 cases and 27 459 controls, including a prior study of fibrotic interstitial pneumonia). Single nucleotide polymorphisms, classical HLA alleles and amino acids were analysed and signals meeting a region-wide association threshold of p<4.5×10-4 and a posterior probability of replication >90% were considered significant. We sought to replicate the previously reported HLA-DQB1 association in the subset of studies independent of the original report. Results: The meta-analysis of all seven studies identified four significant independent single nucleotide polymorphisms associated with IPF risk. However, none met the posterior probability for replication criterion. The HLA-DQB1 association was not replicated in the independent IPF studies. Conclusion: Variation in the HLA region was not consistently associated with risk in studies of IPF. However, this does not preclude the possibility that other genomic regions linked to the immune response may be involved in the aetiology of IPF.

2.
medRxiv ; 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38293162

RESUMEN

Background: Idiopathic pulmonary fibrosis (IPF) is a chronic lung condition that is more prevalent in males than females. The reasons for this are not fully understood, with differing environmental exposures due to historically sex-biased occupations, or diagnostic bias, being possible explanations. To date, over 20 independent genetic variants have been identified to be associated with IPF susceptibility, but these have been discovered when combining males and females. Our aim was to test for the presence of sex-specific associations with IPF susceptibility and assess whether there is a need to consider sex-specific effects when evaluating genetic risk in clinical prediction models for IPF. Methods: We performed genome-wide single nucleotide polymorphism (SNP)-by-sex interaction studies of IPF risk in six independent IPF case-control studies and combined them using inverse-variance weighted fixed effect meta-analysis. In total, 4,561 cases (1,280 females and 2,281 males) and 23,500 controls (8,360 females and 14,528 males) of European genetic ancestry were analysed. We used polygenic risk scores (PRS) to assess differences in genetic risk prediction between males and females. Findings: Three independent genetic association signals were identified. All showed a consistent direction of effect across all individual IPF studies and an opposite direction of effect in IPF susceptibility between females and males. None had been previously identified in IPF susceptibility genome-wide association studies (GWAS). The predictive accuracy of the PRSs were similar between males and females, regardless of whether using combined or sex-specific GWAS results. Interpretation: We prioritised three genetic variants whose effect on IPF risk may be modified by sex, however these require further study. We found no evidence that the predictive accuracy of common SNP-based PRSs varies significantly between males and females.

3.
medRxiv ; 2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37546732

RESUMEN

Introduction: Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial pneumonia marked by progressive lung fibrosis and a poor prognosis. Recent studies have highlighted the potential role of infection in the pathogenesis of IPF and a prior association of the HLA-DQB1 gene with idiopathic fibrotic interstitial pneumonia (including IPF) has been reported. Due to the important role that the Human Leukocyte Antigen (HLA) region plays in the immune response, here we evaluated if HLA genetic variation was associated specifically with IPF risk. Methods: We performed a meta-analysis of associations of the HLA region with IPF risk in individuals of European ancestry from seven independent case-control studies of IPF (comprising a total of 5,159 cases and 27,459 controls, including the prior study of fibrotic interstitial pneumonia). Single nucleotide polymorphisms, classical HLA alleles and amino acids were analysed and signals meeting a region-wide association threshold p<4.5×10-4 and a posterior probability of replication >90% were considered significant. We sought to replicate the previously reported HLA-DQB1 association in the subset of studies independent of the original report. Results: The meta-analysis of all seven studies identified four significant independent single nucleotide polymorphisms associated with IPF risk. However, none met the posterior probability for replication criterion. The HLA-DQB1 association was not replicated in the independent IPF studies. Conclusion: Variation in the HLA region was not consistently associated with risk in studies of IPF. However, this does not preclude the possibility that other genomic regions linked to the immune response may be involved in the aetiology of IPF.

4.
Eur Respir J ; 60(5)2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35798357

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease associated with chronic inflammation and tissue remodelling leading to fibrosis, reduced pulmonary function, respiratory failure and death. Bleomycin (Blm)-induced lung fibrosis in mice replicates several clinical features of human IPF, including prominent lymphoid aggregates of predominantly B-cells that accumulate in the lung adjacent to areas of active fibrosis. We have shown previously a requirement for B-cells in the development of Blm-induced lung fibrosis in mice. To determine the therapeutic potential of inhibiting B-cell function in pulmonary fibrosis, we examined the effects of anti-CD20 B-cell ablation therapy to selectively remove mature B-cells from the immune system and inhibit Blm-induced lung fibrosis. Anti-CD20 B-cell ablation did not reduce fibrosis in this model; however, immune phenotyping of peripheral blood and lung resident cells revealed that anti-CD20-treated mice retained a high frequency of CD19+ CD138+ plasma cells. Interestingly, high levels of CD138+ cells were also identified in the lung tissue of patients with IPF, consistent with the mouse model. Treatment of mice with bortezomib, which depletes plasma cells, reduced the level of Blm-induced lung fibrosis, implicating plasma cells as important effector cells in the development and progression of pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , Enfermedades Pulmonares Intersticiales , Humanos , Ratones , Animales , Bleomicina/farmacología , Células Plasmáticas , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Pulmón/metabolismo , Enfermedades Pulmonares Intersticiales/inducido químicamente
5.
Vet Parasitol ; 309: 109757, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35785612

RESUMEN

We investigated whether measurement of live weight gain (LWG) could be used to deliver targeted selective treatment (TST) with anthelmintic that maintained target live weight (LW) while reducing anthelmintic use. Dairy heifers (n = 546) from four commercial, seasonal calving, pastoral New Zealand dairy herds in the Canterbury region of New Zealand were recruited to compare effects of TST, against suppressive treatment (ST), on LW and LWG in their first year at pasture. Animals were enrolled at weaning (December) and weighed ± anthelmintic treatment every month until May and then in August and September, pre-mating in October. All ST calves were treated every month with an oral anthelmintic at 1 mL/5 kg LW, delivering 0.2 mg abamectin, 8 mg levamisole and 4.5 mg oxfendazole/kg LW, (Control). TST calves received the same anthelmintic at the same time but only if either their individual LWG fell below a pre-specified, time altered, breed specific target for that group (Group target) or if their individual LWG fell below an individual, time altered target for that individual (Individual target). Faecal egg count (FEC) and serum IgA OD levels were recorded from a sample of calves throughout the study. Compared to ST, anthelmintic use was halved in both TST groups (P < 0.001) and there was no evidence for differences in the proportion of calves reaching target LW pre-mating (Control = 80 (95 % CI = 79.1-87.2 %), Group = 78.9 (95 % CI = 59.6-98.2%), Individual = 78.2 (95 % CI = 58.4-97.9 %), P = 0.935). Control calves were heavier pre-mating (310 (95 % CI = 290-330 kg) than Group (300 (95 % CI = 280-320 kg) or Individual (298 (95 % CI = 278-318 kg), P < 0.001). An interaction between FEC and time meant calves with FEC > 200 EPG grew more slowly in the autumn (P < 0.001), but more quickly in the winter (P < 0.001). FEC and IgA OD levels were consistent with levels of parasitism sufficient to impact LWG, but there was no evidence for differences between treatment groups. Sensitivity and specificity of LW and LWG as predictors of reaching target weight varied month-on-month but these results suggest monthly LWG and LW were poor indicators for AHC treatment. In conclusion, TST can be effective in reducing anthelmintic use and maintaining group level performance. LW and LWG sensitivity was 57-85 % and 66-93 %; specificity 38-83 % and 32-79.


Asunto(s)
Antihelmínticos , Enfermedades de los Bovinos , Animales , Antihelmínticos/farmacología , Bovinos , Enfermedades de los Bovinos/tratamiento farmacológico , Heces , Femenino , Inmunoglobulina A , Nueva Zelanda , Recuento de Huevos de Parásitos/veterinaria , Aumento de Peso
6.
Animals (Basel) ; 12(8)2022 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-35454243

RESUMEN

Loline, an alkaloid with several derivatives, has suggested antimicrobial and anthelmintic properties. Therefore, loline was investigated as a natural anthelmintic against Trichostrongylus colubriformis, Teladorsagia circumcincta, and Haemonchus contortus. Preliminary in vitro studies had reduced L3 T. circumcincta establishment but no effect on L3 T. colubriformis larvae migration or H. contortus establishment. While loline-treated lambs had lower establishment of L4 and adult T. circumcincta and L4 T. colubriformis, L4 and adult H. contortus appeared unaffected. Following preliminary study, an in vivo experiment examined lambs infected with a mix of L4 T. circumcincta, T. colubriformis, and adult H. contortus. These lambs were treated with either a loline seed extract (LOL, n = 7), nothing (CON, n = 7), or a non-loline seed extract (NIL, n = 2). There were no differences in worm burdens, fecal egg counts, weight gain, or feed intake between treatments. However, an average growth efficiency (kg LWG/kg DM intake) was detected (p = 0.01) in CON (0.18) which was less than LOL (0.24) or NIL (0.23). There was limited evidence to support an in vivo anti-parasitic effect of loline despite in vitro studies indicating potential benefits. Discrepancies between in vivo and in vitro studies results were potentially a result of loline contact time with larvae, mode of ingestion or the forms of loline present.

7.
Biomedicines ; 9(5)2021 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946612

RESUMEN

The interleukin (IL)-6 family of cytokines and exaggerated signal transducer and activator of transcription (STAT)3 signaling is implicated in idiopathic pulmonary fibrosis (IPF) pathogenesis, but the mechanisms regulating STAT3 expression and function are unknown. Suppressor of cytokine signaling (SOCS)1 and SOCS3 block STAT3, and low SOCS1 levels have been reported in IPF fibroblasts and shown to facilitate collagen production. Fibroblasts and lung tissue from IPF patients and controls were used to examine the mechanisms underlying SOCS1 down-regulation in IPF. A significant reduction in basal SOCS1 mRNA in IPF fibroblasts was confirmed. However, there was no difference in the kinetics of activation, and methylation of SOCS1 in control and IPF lung fibroblasts was low and unaffected by 5'-aza-2'-deoxycytidine' treatment. SOCS1 is a target of microRNA-155 and although microRNA-155 levels were increased in IPF tissue, they were reduced in IPF fibroblasts. Therefore, SOCS1 is not regulated by SOCS1 gene methylation or microRNA155 in these cells. In conclusion, we confirmed that IPF fibroblasts had lower levels of SOCS1 mRNA compared with control fibroblasts, but we were unable to determine the mechanism. Furthermore, although SOCS1 may be important in the fibrotic process, we were unable to find a significant role for SOCS1 in regulating fibroblast function.

8.
Thorax ; 76(1): 73-82, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33214245

RESUMEN

INTRODUCTION: Fibroblastic foci represent the cardinal pathogenic lesion in idiopathic pulmonary fibrosis (IPF) and comprise activated fibroblasts and myofibroblasts, the key effector cells responsible for dysregulated extracellular matrix deposition in multiple fibrotic conditions. The aim of this study was to define the major transcriptional programmes involved in fibrogenesis in IPF by profiling unmanipulated myofibroblasts within fibrotic foci in situ by laser capture microdissection. METHODS: The challenges associated with deriving gene calls from low amounts of RNA and the absence of a meaningful comparator cell type were overcome by adopting novel data mining strategies and by using weighted gene co-expression network analysis (WGCNA), as well as an eigengene-based approach to identify transcriptional signatures, which correlate with fibrillar collagen gene expression. RESULTS: WGCNA identified prominent clusters of genes associated with cell cycle, inflammation/differentiation, translation and cytoskeleton/cell adhesion. Collagen eigengene analysis revealed that transforming growth factor ß1 (TGF-ß1), RhoA kinase and the TSC2/RHEB axis formed major signalling clusters associated with collagen gene expression. Functional studies using CRISPR-Cas9 gene-edited cells demonstrated a key role for the TSC2/RHEB axis in regulating TGF-ß1-induced mechanistic target of rapamycin complex 1 activation and collagen I deposition in mesenchymal cells reflecting IPF and other disease settings, including cancer-associated fibroblasts. CONCLUSION: These data provide strong support for the human tissue-based and bioinformatics approaches adopted to identify critical transcriptional nodes associated with the key pathogenic cell responsible for fibrogenesis in situ and further identify the TSC2/RHEB axis as a potential novel target for interfering with excessive matrix deposition in IPF and other fibrotic conditions.


Asunto(s)
Regulación de la Expresión Génica , Fibrosis Pulmonar Idiopática/genética , ARN/genética , Transcriptoma/genética , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Pulmón/patología , Transducción de Señal , Regulación hacia Arriba
9.
Parasite Immunol ; 42(11): e12776, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32672355

RESUMEN

BACKGROUND AND OBJECTIVES: The possibility of manipulating the immune response in lambs to the gastrointestinal nematode Trichostrongylus colubriformis to reduce production losses associated with infection was investigated. In a series of four experiments, attempts to immunize sheep via the mucosal route to modify the immune response and induce mucosal tolerance are outlined. Initially, a proof of concept study was conducted with lambs being injected with multiple doses of a somatic T colubriformis antigen without an adjuvant in the rectal submucosa and subsequently challenged with T colubriformis L3 larvae. This was followed by a dose-response study comparing different antigen doses to identify the optimum dose of the nematode antigen for successful induction of mucosal tolerance. The final two studies were conducted to determine the larval stage specificity of the parasite antigen and the most suitable site of delivery required to stimulate mucosal tolerance. METHODS: In the proof of concept study, lambs either received repeated injections in the rectal submucosa at 3 × weekly intervals with 15 µg of L3, 11 µg of L4 and 21 µg of immature adult (L5) somatic T colubriformis antigens (ANT) or not (INF) prior to infection with T colubriformis. In the dose-rate study, antigen dose rates of 100%, 50%, 10%, 1% or 0% of the antigen concentration used in the proof of concept study were compared while the larval stage study compared antigen from either L3, L4, L5 stages or combination of all (COMB) and the route of administration study compared antigen delivery into either the rectal submucosa (RE) or sub-cutaneous injection (SC). RESULTS: During infection, lamb growth was improved by antigen treatment between days 21 and 42 in the proof of concept study (P = .009), for groups 10%, 50% and 100% in the dose-rate study (P < .05 for all) and in RE in the route of administration study with no improvement observed in the larval stage study. No differences in faecal egg counts were observed (P > .05 for all). Parasite-specific IgA and IgE showed a dose-response (the dose-rate study), were not affected by larval stage (the larval stage study) and were greater in RE than SC (the route of administration study). IL-4 production following lymphocyte stimulation was greatest in COMB (the larval stage study) and RE (the route of administration study). CONCLUSIONS: Although antigen treatment improved performance, this was inconsistent and appeared to stimulate immunity rather than induce tolerance. Combined larval stages were more efficient than individual stages, and intra-rectal administration was more effective than sub-cutaneous.


Asunto(s)
Anticuerpos Antihelmínticos/sangre , Antígenos Helmínticos/administración & dosificación , Inmunización/veterinaria , Enfermedades de las Ovejas/inmunología , Tricostrongiliasis/veterinaria , Trichostrongylus/inmunología , Animales , Antígenos Helmínticos/inmunología , Desensibilización Inmunológica , Heces/parasitología , Femenino , Tracto Gastrointestinal/parasitología , Inmunidad , Larva , Activación de Linfocitos , Masculino , Ovinos , Enfermedades de las Ovejas/parasitología , Tricostrongiliasis/inmunología , Tricostrongiliasis/parasitología
10.
Am J Respir Crit Care Med ; 201(5): 564-574, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31710517

RESUMEN

Rationale: Idiopathic pulmonary fibrosis (IPF) is a complex lung disease characterized by scarring of the lung that is believed to result from an atypical response to injury of the epithelium. Genome-wide association studies have reported signals of association implicating multiple pathways including host defense, telomere maintenance, signaling, and cell-cell adhesion.Objectives: To improve our understanding of factors that increase IPF susceptibility by identifying previously unreported genetic associations.Methods: We conducted genome-wide analyses across three independent studies and meta-analyzed these results to generate the largest genome-wide association study of IPF to date (2,668 IPF cases and 8,591 controls). We performed replication in two independent studies (1,456 IPF cases and 11,874 controls) and functional analyses (including statistical fine-mapping, investigations into gene expression, and testing for enrichment of IPF susceptibility signals in regulatory regions) to determine putatively causal genes. Polygenic risk scores were used to assess the collective effect of variants not reported as associated with IPF.Measurements and Main Results: We identified and replicated three new genome-wide significant (P < 5 × 10-8) signals of association with IPF susceptibility (associated with altered gene expression of KIF15, MAD1L1, and DEPTOR) and confirmed associations at 11 previously reported loci. Polygenic risk score analyses showed that the combined effect of many thousands of as yet unreported IPF susceptibility variants contribute to IPF susceptibility.Conclusions: The observation that decreased DEPTOR expression associates with increased susceptibility to IPF supports recent studies demonstrating the importance of mTOR signaling in lung fibrosis. New signals of association implicating KIF15 and MAD1L1 suggest a possible role of mitotic spindle-assembly genes in IPF susceptibility.


Asunto(s)
Fibrosis Pulmonar Idiopática/genética , Anciano , Estudios de Casos y Controles , Proteínas de Ciclo Celular/genética , Femenino , Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Cinesinas/genética , Masculino , Persona de Mediana Edad , Medición de Riesgo , Transducción de Señal , Huso Acromático , Serina-Treonina Quinasas TOR/metabolismo
11.
Biochem Biophys Res Commun ; 510(2): 198-204, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30685089

RESUMEN

Malignant mesothelioma is an aggressive fibrous tumor, predominantly of the pleura, with a very poor prognosis. Cell-matrix interactions are recognized important determinants of tumor growth and invasiveness but the role of the extracellular matrix in mesothelioma is unknown. Mesothelioma cells synthesize collagen as well as transforming growth factor-beta (TGF-ß), a key regulator of collagen production. This study examined the effect of inhibiting collagen production on mesothelioma cell proliferation in vitro and tumor growth in vivo. Collagen production by mesothelioma cells was inhibited by incubating cells in vitro with the proline analogue thiaproline (thiazolidine-4-carboxylic acid) or by oral administration of thiaproline in a murine tumor model. Cell cytotoxicity was measured using neutral red uptake and lactate dehydrogenase assays. Proliferation was measured by tritiated thymidine incorporation, and inflammatory cell influx, proliferation, apoptosis and angiogenesis in tumors examined by immunohistochemical labelling. Tumor size was determined by tumor weight and collagen production was measured by HPLC. Thiaproline at non-toxic doses significantly reduced basal and TGF-ß-induced collagen production by over 50% and cell proliferation by over 65%. In vivo thiaproline administration inhibited tumor growth at 10 days, decreasing the median tumor weight by 80%. The mean concentration of collagen was 50% lower in the thiaproline-treated tumors compared with the controls. There were no significant differences in vasculature or inflammatory cell infiltration but apoptosis was increased in thiaproline treated tumors at day 10. In conclusion, these observations strongly support a role for collagen in mesothelioma growth and establish the potential for inhibitors of collagen synthesis in mesothelioma treatment.


Asunto(s)
Colágeno/biosíntesis , Neoplasias Pulmonares/metabolismo , Mesotelioma/metabolismo , Neoplasias Pleurales/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Colágeno/antagonistas & inhibidores , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Femenino , Humanos , Inflamación , Neoplasias Pulmonares/patología , Mesotelioma/patología , Mesotelioma Maligno , Ratones , Ratones Endogámicos CBA , Neoplasias Pleurales/patología , Tiazolidinas/farmacología , Factor de Crecimiento Transformador beta/metabolismo
12.
Int J Mol Sci ; 19(8)2018 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-30103548

RESUMEN

Prostacyclins are extensively used to treat pulmonary arterial hypertension (PAH), a life-threatening disease involving the progressive thickening of small pulmonary arteries. Although these agents are considered to act therapeutically via the prostanoid IP receptor, treprostinil is the only prostacyclin mimetic that potently binds to the prostanoid EP2 receptor, the role of which is unknown in PAH. We hypothesised that EP2 receptors contribute to the anti-proliferative effects of treprostinil in human pulmonary arterial smooth muscle cells (PASMCs), contrasting with selexipag, a non-prostanoid selective IP agonist. Human PASMCs from PAH patients were used to assess prostanoid receptor expression, cell proliferation, and cyclic adenosine monophosphate (cAMP) levels following the addition of agonists, antagonists or EP2 receptor small interfering RNAs (siRNAs). Immunohistochemical staining was performed in lung sections from control and PAH patients. We demonstrate using selective IP (RO1138452) and EP2 (PF-04418948) antagonists that the anti-proliferative actions of treprostinil depend largely on EP2 receptors rather than IP receptors, unlike MRE-269 (selexipag-active metabolite). Likewise, EP2 receptor knockdown selectively reduced the functional responses to treprostinil but not MRE-269. Furthermore, EP2 receptor levels were enhanced in human PASMCs and in lung sections from PAH patients compared to controls. Thus, EP2 receptors represent a novel therapeutic target for treprostinil, highlighting key pharmacological differences between prostacyclin mimetics used in PAH.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Epoprostenol/análogos & derivados , Hipertensión Pulmonar/tratamiento farmacológico , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Adolescente , Adulto , Niño , Epoprostenol/farmacología , Femenino , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Sistemas de Mensajero Secundario/efectos de los fármacos
13.
Thorax ; 73(9): 847-856, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29748250

RESUMEN

INTRODUCTION: Loss of the cystic fibrosis transmembrane conductance regulator in cystic fibrosis (CF) leads to hyperabsorption of sodium and fluid from the airway due to upregulation of the epithelial sodium channel (ENaC). Thickened mucus and depleted airway surface liquid (ASL) then lead to impaired mucociliary clearance. ENaC regulation is thus a promising target for CF therapy. Our aim was to develop siRNA nanocomplexes that mediate effective silencing of airway epithelial ENaC in vitro and in vivo with functional correction of epithelial ion and fluid transport. METHODS: We investigated translocation of nanocomplexes through mucus and their transfection efficiency in primary CF epithelial cells grown at air-liquid interface (ALI).Short interfering RNA (SiRNA)-mediated silencing was examined by quantitative RT-PCR and western analysis of ENaC. Transepithelial potential (Vt), short circuit current (Isc), ASL depth and ciliary beat frequency (CBF) were measured for functional analysis. Inflammation was analysed by histological analysis of normal mouse lung tissue sections. RESULTS: Nanocomplexes translocated more rapidly than siRNA alone through mucus. Transfections of primary CF epithelial cells with nanocomplexes targeting αENaC siRNA, reduced αENaC and ßENaC mRNA by 30%. Transfections reduced Vt, the amiloride-sensitive Isc and mucus protein concentration while increasing ASL depth and CBF to normal levels. A single dose of siRNA in mouse lung silenced ENaC by approximately 30%, which persisted for at least 7 days. Three doses of siRNA increased silencing to approximately 50%. CONCLUSION: Nanoparticle-mediated delivery of ENaCsiRNA to ALI cultures corrected aspects of the mucociliary defect in human CF cells and offers effective delivery and silencing in vivo.


Asunto(s)
Fibrosis Quística/genética , Fibrosis Quística/patología , Canales Epiteliales de Sodio/genética , Silenciador del Gen , ARN Interferente Pequeño , Transfección/métodos , Animales , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Humanos , Ratones , Nanopartículas
14.
Sci Rep ; 8(1): 1906, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382921

RESUMEN

Pulmonary fibrosis is a progressive scarring disorder of the lung with dismal prognosis and no curative therapy. Clusterin, an extracellular chaperone and regulator of cell functions, is reduced in bronchoalveolar lavage fluid of patients with pulmonary fibrosis. However, its distribution and role in normal and fibrotic human lung are incompletely characterized. Immunohistochemical localization of clusterin revealed strong staining associated with fibroblasts in control lung and morphologically normal areas of fibrotic lung but weak or undetectable staining in fibrotic regions and particularly fibroblastic foci. Clusterin also co-localized with elastin in vessel walls and additionally with amorphous elastin deposits in fibrotic lung. Analysis of primary lung fibroblast isolates in vitro confirmed the down-regulation of clusterin expression in fibrotic compared with control lung fibroblasts and further demonstrated that TGF-ß1 is capable of down-regulating fibroblast clusterin expression. shRNA-mediated down-regulation of clusterin did not affect TGF-ß1-induced fibroblast-myofibroblast differentiation but inhibited fibroblast proliferative responses and sensitized to apoptosis. Down-regulation of clusterin in fibrotic lung fibroblasts at least partly due to increased TGF-ß1 may therefore represent an appropriate but insufficient response to limit fibroproliferation. Reduced expression of clusterin in the lung may also limit its extracellular chaperoning activity contributing to dysregulated deposition of extracellular matrix proteins.


Asunto(s)
Clusterina/metabolismo , Sustancias Protectoras/metabolismo , Fibrosis Pulmonar/metabolismo , Líquido del Lavado Bronquioalveolar , Diferenciación Celular/fisiología , Células Cultivadas , Regulación hacia Abajo/fisiología , Elastina/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/metabolismo
15.
Lancet Respir Med ; 5(11): 869-880, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29066090

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with high mortality, uncertain cause, and few treatment options. Studies have identified a significant genetic risk associated with the development of IPF; however, mechanisms by which genetic risk factors promote IPF remain unclear. We aimed to identify genetic variants associated with IPF susceptibility and provide mechanistic insight using gene and protein expression analyses. METHODS: We used a two-stage approach: a genome-wide association study in patients with IPF of European ancestry recruited from nine different centres in the UK and controls selected from UK Biobank (stage 1) matched for age, sex, and smoking status; and a follow-up of associated genetic variants in independent datasets of patients with IPF and controls from two independent US samples from the Chicago consortium and the Colorado consortium (stage 2). We investigated the effect of novel signals on gene expression in large transcriptomic and genomic data resources, and examined expression using lung tissue samples from patients with IPF and controls. FINDINGS: 602 patients with IPF and 3366 controls were selected for stage 1. For stage 2, 2158 patients with IPF and 5195 controls were selected. We identified a novel genome-wide significant signal of association with IPF susceptibility near A-kinase anchoring protein 13 (AKAP13; rs62025270, odds ratio [OR] 1·27 [95% CI 1·18-1·37], p=1·32 × 10-9) and confirmed previously reported signals, including in mucin 5B (MUC5B; rs35705950, OR 2·89 [2·56-3·26], p=1·12 × 10-66) and desmoplakin (DSP; rs2076295, OR 1·44 [1·35-1·54], p=7·81 × 10-28). For rs62025270, the allele A associated with increased susceptibility to IPF was also associated with increased expression of AKAP13 mRNA in lung tissue from patients who had lung resection procedures (n=1111). We showed that AKAP13 is expressed in the alveolar epithelium and lymphoid follicles from patients with IPF, and AKAP13 mRNA expression was 1·42-times higher in lung tissue from patients with IPF (n=46) than that in lung tissue from controls (n=51). INTERPRETATION: AKAP13 is a Rho guanine nucleotide exchange factor regulating activation of RhoA, which is known to be involved in profibrotic signalling pathways. The identification of AKAP13 as a susceptibility gene for IPF increases the prospect of successfully targeting RhoA pathway inhibitors in patients with IPF. FUNDING: UK Medical Research Council, National Heart, Lung, and Blood Institute of the US National Institutes of Health, Agencia Canaria de Investigación, Innovación y Sociedad de la Información, Spain, UK National Institute for Health Research, and the British Lung Foundation.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética , Fibrosis Pulmonar Idiopática/genética , Antígenos de Histocompatibilidad Menor/genética , Proteínas Proto-Oncogénicas/genética , Población Blanca/genética , Anciano , Células Epiteliales Alveolares/metabolismo , Estudios de Casos y Controles , Europa (Continente) , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/fisiología , Transducción de Señal/genética , Estructuras Linfoides Terciarias/genética , Proteína de Unión al GTP rhoA/fisiología
16.
Sci Rep ; 7(1): 700, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28386087

RESUMEN

The inhibition of ENaC may have therapeutic potential in CF airways by reducing sodium hyperabsorption, restoring lung epithelial surface fluid levels, airway hydration and mucociliary function. The challenge has been to deliver siRNA to the lung with sufficient efficacy for a sustained therapeutic effect. We have developed a self-assembling nanocomplex formulation for siRNA delivery to the airways that consists of a liposome (DOTMA/DOPE; L), an epithelial targeting peptide (P) and siRNA (R). LPR formulations were assessed for their ability to silence expression of the transcript of the gene encoding the α-subunit of the sodium channel ENaC in cell lines and primary epithelial cells, in submerged cultures or grown in air-liquid interface conditions. LPRs, containing 50 nM or 100 nM siRNA, showed high levels of silencing, particularly in primary airway epithelial cells. When nebulised these nanocomplexes still retained their biophysical properties and transfection efficiencies. The silencing ability was determined at protein level by confocal microscopy and western blotting. In vivo data demonstrated that these nanoparticles had the ability to silence expression of the α-ENaC subunit gene. In conclusion, these findings show that LPRs can modulate the activity of ENaC and this approach might be promising as co-adjuvant therapy for cystic fibrosis.


Asunto(s)
Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/genética , Técnicas de Transferencia de Gen , Nanopartículas , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transducción Genética , Línea Celular , Células Cultivadas , Fibrosis Quística/genética , Fibrosis Quística/terapia , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Terapia Genética , Liposomas/química , Microscopía Confocal , Péptidos/química , Interferencia de ARN , ARN Interferente Pequeño/química , Transfección
17.
Thorax ; 71(8): 701-11, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27103349

RESUMEN

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is the most rapidly progressive and fatal of all fibrotic conditions with no curative therapies. Common pathomechanisms between IPF and cancer are increasingly recognised, including dysfunctional pan-PI3 kinase (PI3K) signalling as a driver of aberrant proliferative responses. GSK2126458 is a novel, potent, PI3K/mammalian target of rapamycin (mTOR) inhibitor which has recently completed phase I trials in the oncology setting. Our aim was to establish a scientific and dosing framework for PI3K inhibition with this agent in IPF at a clinically developable dose. METHODS: We explored evidence for pathway signalling in IPF lung tissue and examined the potency of GSK2126458 in fibroblast functional assays and precision-cut IPF lung tissue. We further explored the potential of IPF patient-derived bronchoalveolar lavage (BAL) cells to serve as pharmacodynamic biosensors to monitor GSK2126458 target engagement within the lung. RESULTS: We provide evidence for PI3K pathway activation in fibrotic foci, the cardinal lesions in IPF. GSK2126458 inhibited PI3K signalling and functional responses in IPF-derived lung fibroblasts, inhibiting Akt phosphorylation in IPF lung tissue and BAL derived cells with comparable potency. Integration of these data with GSK2126458 pharmacokinetic data from clinical trials in cancer enabled modelling of an optimal dosing regimen for patients with IPF. CONCLUSIONS: Our data define PI3K as a promising therapeutic target in IPF and provide a scientific and dosing framework for progressing GSK2126458 to clinical testing in this disease setting. A proof-of-mechanism trial of this agent is currently underway. TRIAL REGISTRATION NUMBER: NCT01725139, pre-clinical.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinolinas/uso terapéutico , Sulfonamidas/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proliferación Celular , Ensayos Clínicos como Asunto , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/patología , Piridazinas , Transducción de Señal , Resultado del Tratamiento
18.
Sci Rep ; 6: 23125, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26975732

RESUMEN

Gene therapy for cystic fibrosis using non-viral, plasmid-based formulations has been the subject of intensive research for over two decades but a clinically viable product has yet to materialise in large part due to inefficient transgene expression. Minicircle DNA give enhanced and more persistent transgene expression compared to plasmid DNA in a number of organ systems but has not been assessed in the lung. In this study we compared minicircle DNA with plasmid DNA in transfections of airway epithelial cells. In vitro, luciferase gene expression from minicircles was 5-10-fold higher than with plasmid DNA. In eGFP transfections in vitro both the mean fluorescence intensity and percentage of cells transfected was 2-4-fold higher with minicircle DNA. Administration of equimolar amounts of DNA to mouse lungs resulted in a reduced inflammatory response and more persistent transgene expression, with luciferase activity persisting for 2 weeks from minicircle DNA compared to plasmid formulations. Transfection of equal mass amounts of DNA in mouse lungs resulted in a 6-fold increase in transgene expression in addition to more persistent transgene expression. Our findings have clear implications for gene therapy of airway disorders where plasmid DNA transfections have so far proven inefficient in clinical trials.


Asunto(s)
Dineínas Axonemales/genética , ADN Circular/genética , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Luciferasas de Luciérnaga/genética , Pulmón/metabolismo , Animales , Dineínas Axonemales/metabolismo , Línea Celular , Citocinas/metabolismo , Células Epiteliales/metabolismo , Femenino , Expresión Génica , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Luciferasas de Luciérnaga/metabolismo , Ratones , Plásmidos , Transfección , Transgenes
19.
Thorax ; 71(6): 565-7, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26911575

RESUMEN

Caffeine is a commonly used food additive found naturally in many products. In addition to potently stimulating the central nervous system caffeine is able to affect various systems within the body including the cardiovascular and respiratory systems. Importantly, caffeine is used clinically to treat apnoea and bronchopulmonary dysplasia in premature babies. Recently, caffeine has been shown to exhibit antifibrotic effects in the liver in part through reducing collagen expression and deposition, and reducing expression of the profibrotic cytokine TGFß. The potential antifibrotic effects of caffeine in the lung have not previously been investigated. Using a combined in vitro and ex vivo approach we have demonstrated that caffeine can act as an antifibrotic agent in the lung by acting on two distinct cell types, namely epithelial cells and fibroblasts. Caffeine inhibited TGFß activation by lung epithelial cells in a concentration-dependent manner but had no effect on TGFß activation in fibroblasts. Importantly, however, caffeine abrogated profibrotic responses to TGFß in lung fibroblasts. It inhibited basal expression of the α-smooth muscle actin gene and reduced TGFß-induced increases in profibrotic genes. Finally, caffeine reduced established bleomycin-induced fibrosis after 5 days treatment in an ex vivo precision-cut lung slice model. Together, these findings suggest that there is merit in further investigating the potential use of caffeine, or its analogues, as antifibrotic agents in the lung.


Asunto(s)
Cafeína/farmacología , Células Epiteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Fibrosis Pulmonar/genética , Transducción de Señal/efectos de los fármacos
20.
Clin Sci (Lond) ; 130(8): 575-86, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26744410

RESUMEN

Fibroblasts derived from the lungs of patients with idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc) produce low levels of prostaglandin (PG) E2, due to a limited capacity to up-regulate cyclooxygenase-2 (COX-2). This deficiency contributes functionally to the fibroproliferative state, however the mechanisms responsible are incompletely understood. In the present study, we examined whether the reduced level of COX-2 mRNA expression observed in fibrotic lung fibroblasts is regulated epigenetically. The DNA methylation inhibitor, 5-aza-2'-deoxycytidine (5AZA) restored COX-2 mRNA expression by fibrotic lung fibroblasts dose dependently. Functionally, this resulted in normalization of fibroblast phenotype in terms of PGE2 production, collagen mRNA expression and sensitivity to apoptosis. COX-2 methylation assessed by bisulfite sequencing and methylation microarrays was not different in fibrotic fibroblasts compared with controls. However, further analysis of the methylation array data identified a transcriptional regulator, chromosome 8 open reading frame 4 (thyroid cancer protein 1, TC-1) (c8orf4), which is hypermethylated and down-regulated in fibrotic fibroblasts compared with controls. siRNA knockdown of c8orf4 in control fibroblasts down-regulated COX-2 and PGE2 production generating a phenotype similar to that observed in fibrotic lung fibroblasts. Chromatin immunoprecipitation demonstrated that c8orf4 regulates COX-2 expression in lung fibroblasts through binding of the proximal promoter. We conclude that the decreased capacity of fibrotic lung fibroblasts to up-regulate COX-2 expression and COX-2-derived PGE2 synthesis is due to an indirect epigenetic mechanism involving hypermethylation of the transcriptional regulator, c8orf4.


Asunto(s)
Ciclooxigenasa 2/genética , Metilación de ADN , Epigénesis Genética , Fibroblastos/enzimología , Pulmón/enzimología , Proteínas de Neoplasias/genética , Fibrosis Pulmonar/genética , Esclerodermia Sistémica/genética , Anciano , Sitios de Unión , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/antagonistas & inhibidores , Metilasas de Modificación del ADN/metabolismo , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/patología , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esclerodermia Sistémica/enzimología , Esclerodermia Sistémica/patología , Transcripción Genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA