Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Respir Res ; 24(1): 217, 2023 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-37674160

RESUMEN

Cystic fibrosis (CF) is caused by defects of the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CFTR-modulating drugs may overcome specific defects, such as the case of Trikafta, which is a clinically approved triple combination of Elexacaftor, Tezacaftor and Ivacaftor (ETI) that exhibited a strong ability to rescue the function of the most frequent F508del pathogenic variant even in genotypes with the mutated allele in single copy. Nevertheless, most rare genotypes lacking the F508del allele are still not eligible for targeted therapies. Via the innovative approach of using nasal conditionally reprogrammed cell (CRC) cell-based models that mimic patient disease in vitro, which are obtainable from each patient due to the 100% efficiency of the cell culture establishment, we theratyped orphan CFTR mutation L1077P. Protein studies, Forskolin-induced organoid swelling, and Ussing chamber assays congruently proved the L1077P variant function rescue by ETI. Notably, this rescue takes place even in the context of a single-copy L1077P allele, which appears to enhance its expression. Thus, the possibility of single-allele treatment also arises for rare genotypes, with an allele-specific modulation as part of the mechanism. Of note, besides providing indication of drug efficacy with respect to specific CFTR pathogenic variants or genotypes, this approach allows the evaluation of the response of single-patient cells within their genetic background. In this view, our studies support in vitro guided personalized CF therapies also for rare patients who are nearly excluded from clinical trials.


Asunto(s)
Fibrosis Quística , Humanos , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética
2.
Int J Mol Sci ; 24(12)2023 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-37373505

RESUMEN

Despite the promising results of new CFTR targeting drugs designed for the recovery of F508del- and class III variants activity, none of them have been approved for individuals with selected rare mutations, because uncharacterized CFTR variants lack information associated with the ability of these compounds in recovering their molecular defects. Here we used both rectal organoids (colonoids) and primary nasal brushed cells (hNEC) derived from a CF patient homozygous for A559T (c.1675G>A) variant to evaluate the responsiveness of this pathogenic variant to available CFTR targeted drugs that include VX-770, VX-809, VX-661 and VX-661 combined with VX-445. A559T is a rare mutation, found in African-Americans people with CF (PwCF) with only 85 patients registered in the CFTR2 database. At present, there is no treatment approved by FDA (U.S. Food and Drug Administration) for this genotype. Short-circuit current (Isc) measurements indicate that A559T-CFTR presents a minimal function. The acute addition of VX-770 following CFTR activation by forskolin had no significant increment of baseline level of anion transport in both colonoids and nasal cells. However, the combined treatment, VX-661-VX-445, significantly increases the chloride secretion in A559T-colonoids monolayers and hNEC, reaching approximately 10% of WT-CFTR function. These results were confirmed by forskolin-induced swelling assay and by western blotting in rectal organoids. Overall, our data show a relevant response to VX-661-VX-445 in rectal organoids and hNEC with CFTR genotype A559T/A559T. This could provide a strong rationale for treating patients carrying this variant with VX-661-VX-445-VX-770 combination.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Humanos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Colforsina/uso terapéutico , Benzodioxoles/farmacología , Mutación , Organoides , Genotipo
3.
EMBO Rep ; 19(7)2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29752334

RESUMEN

Heat-shock factor 1 (HSF1) is the master transcription factor that regulates the response to proteotoxic stress by controlling the transcription of many stress-responsive genes including the heat-shock proteins. Here, we show a novel molecular mechanism controlling the activation of HSF1. We demonstrate that transglutaminase type 2 (TG2), dependent on its protein disulphide isomerase activity, triggers the trimerization and activation of HSF1 regulating adaptation to stress and proteostasis impairment. In particular, we find that TG2 loss of function correlates with a defect in the nuclear translocation of HSF1 and in its DNA-binding ability to the HSP70 promoter. We show that the inhibition of TG2 restores the unbalance in HSF1-HSP70 pathway in cystic fibrosis (CF), a human disorder characterized by deregulation of proteostasis. The absence of TG2 leads to an increase of about 40% in CFTR function in a new experimental CF mouse model lacking TG2. Altogether, these results indicate that TG2 plays a key role in the regulation of cellular proteostasis under stressful cellular conditions through the modulation of the heat-shock response.


Asunto(s)
Fibrosis Quística/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al GTP/genética , Factores de Transcripción del Choque Térmico/genética , Transglutaminasas/genética , Animales , Fibrosis Quística/patología , Regulación de la Expresión Génica , Respuesta al Choque Térmico/genética , Humanos , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Proteína Disulfuro Isomerasas/genética , Proteína Glutamina Gamma Glutamiltransferasa 2 , Procesamiento Proteico-Postraduccional/genética , Proteostasis/genética , Transducción de Señal
5.
Clin Chim Acta ; 558: 118317, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38580140

RESUMEN

Cystic fibrosis (CF) is a life-limiting genetic disorder characterized by defective chloride ion transport due to mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Early detection through newborn screening programs significantly improves outcomes for individuals with CF by enabling timely intervention. Here, we report the identification of an Alu element insertion within the exon 15 of CFTR gene, initially overlooked in standard next-generation sequencing analyses. However, using traditional molecular techniques, based on polymerase chain reaction and Sanger sequencing, allowed the identification of the Alu element and the reporting of a correct diagnosis. Our analysis, based on bioinformatics tools and molecular techniques, revealed that the Alu element insertion severely affects the gene expression, splicing patterns, and structure of CFTR protein. In conclusion, this study emphasizes the importance of how the integration of human expertise and modern technologies represents a pivotal step forward in genomic medicine, ensuring the delivery of precision healthcare to individuals affected by genetic diseases.


Asunto(s)
Elementos Alu , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Pruebas Genéticas , Humanos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Elementos Alu/genética , Fibrosis Quística/genética , Fibrosis Quística/diagnóstico , Pruebas Genéticas/métodos , Recién Nacido , Masculino , Femenino
6.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38719750

RESUMEN

Celiac disease (CD) is an autoimmune enteropathy resulting from an interaction between diet, genome, and immunity. Although many patients respond to a gluten-free diet, in a substantive number of individuals, the intestinal injury persists. Thus, other factors might amplify the ongoing inflammation. Candida albicans is a commensal fungus that is well adapted to the intestinal life. However, specific conditions increase Candida pathogenicity. The hypothesis that Candida may be a trigger in CD has been proposed after the observation of similarity between a fungal wall component and two CD-related gliadin T-cell epitopes. However, despite being implicated in intestinal disorders, Candida may also protect against immune pathologies highlighting a more intriguing role in the gut. Herein, we postulated that a state of chronic inflammation associated with microbial dysbiosis and leaky gut are favorable conditions that promote C. albicans pathogenicity eventually contributing to CD pathology via a mast cells (MC)-IL-9 axis. However, the restoration of immune and microbial homeostasis promotes a beneficial C. albicans-MC cross-talk favoring the attenuation of CD pathology to alleviate CD pathology and symptoms.


Asunto(s)
Candida albicans , Enfermedad Celíaca , Homeostasis , Mastocitos , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/metabolismo , Humanos , Candida albicans/patogenicidad , Candida albicans/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Microbioma Gastrointestinal/inmunología , Disbiosis/inmunología , Candidiasis/inmunología , Candidiasis/microbiología , Animales , Candida/patogenicidad , Candida/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo
7.
J R Soc Interface ; 20(199): 20220719, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36872917

RESUMEN

Coordinated rotational motion is an intriguing, yet still elusive mode of collective cell migration, which is relevant in pathological and morphogenetic processes. Most of the studies on this topic have been carried out on epithelial cells plated on micropatterned substrates, where cell motion is confined in regions of well-defined shapes coated with extracellular matrix adhesive proteins. The driver of collective rotation in such conditions has not been clearly elucidated, although it has been speculated that spatial confinement can play an essential role in triggering cell rotation. Here, we study the growth of epithelial cell colonies freely expanding (i.e. with no physical constraints) on the surface of cell culture plates and focus on collective cell rotation in such conditions, a case which has received scarce attention in the literature. One of the main findings of our work is that coordinated cell rotation spontaneously occurs in cell clusters in the free growth regime, thus implying that cell confinement is not necessary to elicit collective rotation as previously suggested. The extent of collective rotation was size and shape dependent: a highly coordinated disc-like rotation was found in small cell clusters with a round shape, while collective rotation was suppressed in large irregular cell clusters generated by merging of different clusters in the course of their growth. The angular motion was persistent in the same direction, although clockwise and anticlockwise rotations were equally likely to occur among different cell clusters. Radial cell velocity was quite low as compared to the angular velocity, in agreement with the free expansion regime where cluster growth is essentially governed by cell proliferation. A clear difference in morphology was observed between cells at the periphery and the ones in the core of the clusters, the former being more elongated and spread out as compared to the latter. Overall, our results, to our knowledge, provide the first quantitative and systematic evidence that coordinated cell rotation does not require a spatial confinement and occurs spontaneously in freely expanding epithelial cell colonies, possibly as a mechanism for the system.


Asunto(s)
Técnicas de Cultivo de Célula , Células Epiteliales , Movimiento Celular , Proliferación Celular , Proteínas de la Matriz Extracelular
8.
Biosensors (Basel) ; 13(4)2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37185523

RESUMEN

Transglutaminase 2 (TG2) is a ubiquitously expressed member of the transglutaminase family with Ca2+-dependent protein crosslinking activity. Its subcellular localization is crucial in determining its function, and indeed, TG2 is found in the extracellular matrix, mitochondria, recycling endosomes, plasma membrane, cytosol, and nucleus because it is associated with cell growth, differentiation, and apoptosis. It is involved in several pathologies, such as celiac disease, cardiovascular, hepatic, renal, and fibrosis diseases, carrying out opposite functions of up and down regulation in the progression of the same pathology. Therefore, this fine regulation requires a very sensitive and specific method of identification of TG2, which is to be detected in very small quantities in a deregulated condition. Here, we demonstrate the possibility of detecting TG2 down to attomolar concentration by using organic electrochemical transistors driven by gold electrodes functionalized with anti-TG2 antibodies. In particular, a direct correlation between the TG2 concentration and the transistor transconductance values, as extracted from typical transfer curves, was found. Overall, our findings highlight the potentialities of this new biosensing approach for the detection of TG2 in the context of pathological diseases, offering a rapid and cost-effective alternative to traditional methods.


Asunto(s)
Proteínas de Unión al GTP , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteínas de Unión al GTP/metabolismo , Transglutaminasas/metabolismo , Hígado , Apoptosis
9.
Biosensors (Basel) ; 13(7)2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37504137

RESUMEN

The global COVID-19 pandemic has had severe consequences from the social and economic perspectives, compelling the scientific community to focus on the development of effective diagnostics that can combine a fast response and accurate sensitivity/specificity performance. Presently available commercial antigen-detecting rapid diagnostic tests (Ag-RDTs) are very fast, but still face significant criticisms, mainly related to their inability to amplify the protein signal. This translates to a limited sensitive outcome and, hence, a reduced ability to hamper the spread of SARS-CoV-2 infection. To answer the urgent need for novel platforms for the early, specific and highly sensitive detection of the virus, this paper deals with the use of organic electrochemical transistors (OECTs) as very efficient ion-electron converters and amplifiers for the detection of spike proteins and their femtomolar concentration. The electrical response of the investigated OECTs was carefully analyzed, and the changes in the parameters associated with the transconductance (i.e., the slope of the transfer curves) in the gate voltage range between 0 and 0.3 V were found to be more clearly correlated with the spike protein concentration. Moreover, the functionalization of OECT-based biosensors with anti-spike and anti-nucleocapside proteins, the major proteins involved in the disease, demonstrated the specificity of these devices, whose potentialities should also be considered in light of the recent upsurge of the so-called "long COVID" syndrome.


Asunto(s)
Técnicas Biosensibles , COVID-19 , Humanos , COVID-19/diagnóstico , Glicoproteína de la Espiga del Coronavirus , Pandemias , SARS-CoV-2 , Transistores Electrónicos , Proteínas
10.
Front Immunol ; 14: 1093212, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36923406

RESUMEN

Cystic fibrosis (CF) is a rare autosomal recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The most common mutation is F508del-CFTR (ΔF) which leads the encoded ion channel towards misfolding and premature degradation. The disease is characterized by chronic bronchopulmonary obstruction, inflammation and airways colonization by bacteria, which are the major cause of morbidity and mortality. The STING pathway is the main signaling route activated in the presence of both self and pathogen DNA, leading to Type I Interferon (IFN I) production and the innate immune response. In this study, we show for the first time the relationship existing in CF between resistant and recurrent opportunistic infections by Pseudomonas aeruginosa and the innate immunity impairment. We demonstrate through ex vivo and in vivo experiments that the pathway is inadequately activated in ΔF condition and the use of direct STING agonists, as 2',3'-cyclic GMP-AMP (2', 3' cGAMP), is able to restore the immune response against bacterial colonization. Indeed, upon treatment with the STING pathway agonists, we found a reduction of colony forming units (CFUs) consequent to IFN-ß enhanced production in Pseudomonas aeruginosa infected bone marrow derived macrophages and lung tissues from mice affected by Cystic Fibrosis. Importantly, we also verified that the impairment detected in the primary PBMCs obtained from ΔF patients can be corrected by 2', 3' cGAMP. Our work indicates that the cGAS/STING pathway integrity is crucial in the Cystic Fibrosis response against pathogens and that the restoration of the pathway by 2', 3' cGAMP could be exploited as a possible new target for the symptomatic treatment of the disease.


Asunto(s)
Fibrosis Quística , Interferón Tipo I , Ratones , Animales , Fibrosis Quística/microbiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Inmunidad Innata/genética , Interferón Tipo I/metabolismo , Macrófagos , Proteínas Serina-Treonina Quinasas/metabolismo , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo
11.
iScience ; 26(11): 108180, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38026150

RESUMEN

Mutation targeted therapy in cystic fibrosis (CF) is still not eligible for all CF subjects, especially for cases carrying rare variants such as the CFTR genotype W57G/A234D (c.169T>G/c.701C>A). We performed in silico analysis of the effects of these variants on protein stability, which we functionally characterized using colonoids and reprogrammed nasal epithelial cells. The effect of mutations on cystic fibrosis transmembrane conductance regulator (CFTR) protein was analyzed by western blotting, forskolin-induced swelling (FIS), and Ussing chamber analysis. We detected a residual CFTR function that increases following treatment with the CFTR modulators VX661±VX445±VX770, correlates among models, and is associated with increased CFTR protein levels following treatment with CFTR correctors. In vivo treatment with VX770 reduced sweat chloride concentration to non-CF levels, increased the number of CFTR-dependent sweat droplets, and induced a 6% absolute increase in predicted FEV1% after 27 weeks of treatment indicating the relevance of theratyping with patient-derived cells in CF.

12.
Pediatr Res ; 71(5): 532-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22314661

RESUMEN

INTRODUCTION: [corrected] K562(S) agglutination has been used as a rapid and economic tool for the in vitro screening of the toxicity of cereal fractions and prolamins in celiac disease (CD). A strict correlation has been reported between the toxicity of cereals and cereal fractions for celiac patients and their ability to agglutinate K562(S) cells. Whether this specificity of K562(S)-cell agglutination is caused by the activation of the same pathogenic events triggered by toxic cereal fractions in CD intestine or simply represents a bystander event of gluten toxicity is, however, unknown. METHODS: K562(S) cells were incubated in vitro with the peptic-tryptic digest of wheat gliadin. RESULTS: The agglutination of K562(S) cells by wheat gliadin peptides is orchestrated by a cascade of very early events occurring at the K562(S)-cell surface similar to those occurring at the intestinal epithelial surface. They involve a rapid increase in intracellular calcium levels that activate tissue transglutaminase (TG2), leading to a rapid actin reorganization that is pivotal in driving cell agglutination. These specific effects of toxic cereals are phenocopied by the gliadin-derived peptide p31-43, which orchestrates the activation of innate response to gliadin in CD. DISCUSSION: Our study provides the rationale for the extensive use of K562(S)-cell agglutination as a valuable tool for screening cereal toxicity.


Asunto(s)
Adhesión Celular , Gliadina/metabolismo , Transglutaminasas/metabolismo , Activación Enzimática , Proteínas de Unión al GTP , Humanos , Células K562 , Proteína Glutamina Gamma Glutamiltransferasa 2
13.
J Immunol ; 183(4): 2775-84, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19625650

RESUMEN

Cystic fibrosis (CF) is a monogenic disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. CF is characterized by chronic bacterial lung infections and inflammation, and we have previously reported that tissue transglutaminase (TG2), a multifunctional enzyme critical to several diseases, is constitutively up-regulated in CF airways and drives chronic inflammation. Here, we demonstrate that the generation of an oxidative stress induced by CFTR-defective function leads to protein inhibitor of activated STAT (PIAS)y-mediated TG2 SUMOylation and inhibits TG2 ubiquitination and proteasome degradation, leading to sustained TG2 activation. This prevents peroxisome proliferator-activated receptor (PPAR)gamma and IkBalpha SUMOylation, leading to NF-kappaB activation and to an uncontrolled inflammatory response. Cellular homeostasis can be restored by small ubiquitin-like modifier (SUMO)-1 or PIASy gene silencing, which induce TG2 ubiquitination and proteasome degradation, restore PPARgamma SUMOylation, and prevent IkBalpha cross-linking and degradation, thus switching off inflammation. Manganese superoxide dismutase overexpression as well as the treatment with the synthetic superoxide dismutase mimetic EUK-134 control PIASy-TG2 interaction and TG2 SUMOylation. TG2 inhibition switches off inflammation in vitro as well as in vivo in a homozygous F508del-CFTR mouse model. Thus, TG2 may function as a link between oxidative stress and inflammation by driving the decision as to whether a protein should undergo SUMO-mediated regulation or degradation. Targeting TG2-SUMO interactions might represent a new option to control disease evolution in CF patients as well as in other chronic inflammatory diseases, neurodegenerative pathologies, and cancer.


Asunto(s)
Mediadores de Inflamación/metabolismo , Estrés Oxidativo/inmunología , Proteína SUMO-1/metabolismo , Transglutaminasas/metabolismo , Animales , Línea Celular Tumoral , Fibrosis Quística/enzimología , Fibrosis Quística/inmunología , Fibrosis Quística/patología , Modelos Animales de Enfermedad , Femenino , Proteínas de Unión al GTP , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Ratones , Ratones Mutantes , Proteínas de Unión a Poli-ADP-Ribosa , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteínas Inhibidoras de STAT Activados/fisiología , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/enzimología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Transglutaminasas/antagonistas & inhibidores , Transglutaminasas/fisiología
14.
Gut ; 59(3): 311-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19951908

RESUMEN

BACKGROUND: An unresolved question in coeliac disease is to understand how some toxic gliadin peptides, in particular p31-43, can initiate an innate response and lead to tissue transglutaminase (TG2) upregulation in coeliac intestine and gliadin sensitive epithelial cell lines. Aim We addressed whether the epithelial uptake of p31-43 induces an intracellular pro-oxidative envoronment favouring TG2 activation and leading to the innate immune response. METHODS: The time course of intracellular delivery to lysosomes of p31-43, palpha-2 or palpha-9 gliadin peptides was analysed in T84 and Caco-2 epithelial cells. The effects of peptide challenge on oxidative stress, TG2 and peroxisome proliferator-activated receptor (PPAR)gamma ubiquitination and p42/44-mitogen activated protein (MAP) kinase or tyrosine phosphorylation were investigated in cell lines and cultured coeliac disease biopsies with/without anti-oxidant treatment or TG2 gene silencing by immunoprecipitation, western blot, confocal microscopy and Fluorenscence Transfer Resonance Energy (FRET) analysis. RESULTS: After 24 h of challenge p31-43, but not palpha-2 or palpha-9, is still retained within LAMP1-positive perinuclear vesicles and leads to increased levels of reactive oxygen species (ROS) that inhibit TG2 ubiquitination and lead to increases of TG2 protein levels and activation. TG2 induces cross-linking, ubiquitination and proteasome degradation of PPARgamma. Treatment with the antioxidant EUK-134 as well as TG2 gene silencing restored PPARgamma levels and reversed all monitored signs of innate activation, as indicated by the dramatic reduction of tyrosine and p42/p44 phosphorylation. CONCLUSION: p31-43 accumulation in lysosomes leads to epithelial activation via the ROS-TG2 axis. TG2 works as a rheostat of ubiquitination and proteasome degradation and drives inflammation via PPARgamma downregulation.


Asunto(s)
Enfermedad Celíaca/metabolismo , Gliadina/metabolismo , Lisosomas/metabolismo , PPAR gamma/metabolismo , Fragmentos de Péptidos/metabolismo , Transglutaminasas/fisiología , Adolescente , Adulto , Regulación hacia Abajo/fisiología , Células Epiteliales/metabolismo , Proteínas de Unión al GTP , Humanos , Mucosa Intestinal/metabolismo , Técnicas de Cultivo de Órganos , Estrés Oxidativo/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Adulto Joven
15.
PLoS One ; 16(1): e0245302, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33412572

RESUMEN

This study aims to investigate cholesterol metabolism in a mouse model with cystic fibrosis (CF) by the comparison of affected homozygous versus wild type (WT) mice. In particular, we evaluated the effects of a diet enriched with cholesterol in both mice groups in comparison with the normal diet. To this purpose, beyond serum and liver cholesterol, we analyzed serum phytosterols as indirect markers of intestinal absorption of cholesterol, liver lathosterol as indirect marker of de novo cholesterol synthesis, liver cholestanol (a catabolite of bile salts synthesis) and the liver mRNA levels of LDL receptor (LDLR), 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CoAR), acyl CoA:cholesterol acyl transferase 2 (ACAT2), cytochrome P450 7A1 (CYP7A1) and tumor necrosis factor alpha (TNFα). CF mice showed lower intestinal absorption and higher liver synthesis of cholesterol than WT mice. In WT mice, the cholesterol supplementation inhibits the synthesis of liver cholesterol and enhances its catabolism, while in CF mice we did not observe a reduction of LDLR and HMG-CoAR expression (probably due to an altered feed-back), causing an increase of intracellular cholesterol. In addition, we observed a further increase (5-fold) in TNFα mRNA levels. This preliminary study suggests that in CF mice there is a vicious circle in which the altered synthesis/secretion of bile salts may reduce the digestion/absorption of cholesterol. As a result, the liver increases the biosynthesis of cholesterol that accumulates in the cells, triggering inflammation and further compromising the metabolism of bile salts.


Asunto(s)
Colesterol/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/patología , Metabolismo de los Lípidos , Hígado/metabolismo , Mutación , Esteroide Hidroxilasas/metabolismo , Animales , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Femenino , Homocigoto , Masculino , Tasa de Depuración Metabólica , Ratones
16.
Nutrients ; 13(4)2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33917155

RESUMEN

Exposure to gluten, a protein present in wheat rye and barley, is the major inducer for human Celiac Disease (CD), a chronic autoimmune enteropathy. CD occurs in about 1% worldwide population, in genetically predisposed individuals bearing human leukocyte antigen (HLA) DQ2/DQ8. Gut epithelial cell stress and the innate immune activation are responsible for the breaking oral tolerance to gliadin, a gluten component. To date, the only treatment available for CD is a long-term gluten-free diet. Several studies have shown that an altered composition of the intestinal microbiota (dysbiosis) could play a key role in the pathogenesis of CD through the modulation of intestinal permeability and the regulation of the immune system. Here, we show that gliadin induces a chronic endoplasmic reticulum (ER) stress condition in the small intestine of a gluten-sensitive mouse model and that the coadministration of probiotics efficiently attenuates both the unfolded protein response (UPR) and gut inflammation. Moreover, the composition of probiotics formulations might differ in their activity at molecular level, especially toward the three axes of the UPR. Therefore, probiotics administration might potentially represent a new valuable strategy to treat gluten-sensitive patients, such as those affected by CD.


Asunto(s)
Suplementos Dietéticos , Estrés del Retículo Endoplásmico , Intolerancia Alimentaria/terapia , Tracto Gastrointestinal/patología , Gliadina/efectos adversos , Glútenes/efectos adversos , Inflamación/patología , Probióticos/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Células CACO-2 , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Permeabilidad , Probióticos/administración & dosificación , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas/metabolismo , Regulación hacia Arriba
17.
Oncotarget ; 10(43): 4492-4500, 2019 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-31321000

RESUMEN

Cystic Fibrosis (CF) is the most frequent lethal monogenetic disease affecting humans. CF is characterized by mutations in cystic fibrosis transmembrane conductance regulator (CFTR), a chloride channel whose malfunction triggers the activation of transglutaminase-2 (TGM2), as well as the inactivation of the Beclin-1 (BECN1) complex resulting in disabled autophagy. CFTR inhibition, TGM2 activation and BECN1 sequestration engage in an 'infernal trio' that locks the cell in a pro-inflammatory state through anti-homeostatic feedforward loops. Thus, stimulation of CFTR function, TGM2 inhibition and autophagy stimulation can be used to treat CF patients. Several studies indicate that patients with CF have a higher incidence of celiac disease (CD) and that mice bearing genetically determined CFTR defects are particularly sensitive to the enteropathogenic effects of the orally supplied gliadin (a gluten-derived protein). A gluten/gliadin-derived peptide (P31-43) inhibits CFTR in mouse intestinal epithelial cells, causing a local stress response that contributes to the immunopathology of CD. In particular, P31-43-induced CFTR inhibition elicits an epithelial stress response perturbing proteostasis. This event triggers TGM2 activation, BECN1 sequestration and results in molecular crosslinking of CFTR and P31-43 by TGM2. Importantly, stimulation of CFTR function with a pharmacological potentiator (Ivacaftor), which is approved for the treatment of CF, could attenuate the autophagy-inhibition and pro-inflammatory effects of gliadin in preclinical models of CD. Thus, CD shares with CF a common molecular mechanism involving CFTR inhibition that might respond to drugs that intercept the "infernal trio". Here, we highlight how drugs available for CF treatment could be repurposed for the therapy of CD.

18.
Aging (Albany NY) ; 11(7): 2003-2019, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30981209

RESUMEN

In celiac disease (CD), an intolerance to dietary gluten/gliadin, antigenic gliadin peptides trigger an HLA-DQ2/DQ8-restricted adaptive Th1 immune response. Epithelial stress, induced by other non-antigenic gliadin peptides, is required for gliadin to become fully immunogenic. We found that cystic-fibrosis-transmembrane-conductance-regulator (CFTR) acts as membrane receptor for gliadin-derived peptide P31-43, as it binds to CFTR and impairs its channel function. P31-43-induced CFTR malfunction generates epithelial stress and intestinal inflammation. Maintaining CFTR in an active open conformation by the CFTR potentiators VX-770 (Ivacaftor) or Vrx-532, prevents P31-43 binding to CFTR and controls gliadin-induced manifestations. Here, we evaluated the possibility that the over-the-counter nutraceutical genistein, known to potentiate CFTR function, would allow to control gliadin-induced alterations. We demonstrated that pre-treatment with genistein prevented P31-43-induced CFTR malfunction and an epithelial stress response in Caco-2 cells. These effects were abrogated when the CFTR gene was knocked out by CRISP/Cas9 technology, indicating that genistein protects intestinal epithelial cells by potentiating CFTR function. Notably, genistein protected gliadin-sensitive mice from intestinal CFTR malfunction and gliadin-induced inflammation as it prevented gliadin-induced IFN-γ production by celiac peripheral-blood-mononuclear-cells (PBMC) cultured ex-vivo in the presence of P31-43-challenged Caco-2 cells. Our results indicate that natural compounds capable to increase CFTR channel gating might be used for the treatment of CD.


Asunto(s)
Enfermedad Celíaca/prevención & control , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Genisteína/farmacología , Gliadina/toxicidad , Fragmentos de Péptidos/toxicidad , Animales , Células CACO-2 , Enfermedad Celíaca/etiología , Enfermedad Celíaca/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Gliadina/inmunología , Humanos , Interferón gamma/biosíntesis , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Fragmentos de Péptidos/inmunología , Unión Proteica
20.
Cell Rep ; 23(6): 1767-1778, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29742432

RESUMEN

Candida albicans is implicated in intestinal diseases. Identifying host signatures that discriminate between the pathogenic versus commensal nature of this human commensal is clinically relevant. In the present study, we identify IL-9 and mast cells (MCs) as key players of Candida commensalism and pathogenicity. By inducing TGF-ß in stromal MCs, IL-9 pivotally contributes to mucosal immune tolerance via the indoleamine 2,3-dioxygenase enzyme. However, Candida-driven IL-9 and mucosal MCs also contribute to barrier function loss, dissemination, and inflammation in experimental leaky gut models and are upregulated in patients with celiac disease. Inflammatory dysbiosis occurs with IL-9 and MC deficiency, indicating that the activity of IL-9 and MCs may go beyond host immunity to include regulation of the microbiota. Thus, the output of the IL-9/MC axis is highly contextual during Candida colonization and reveals how host immunity and the microbiota finely tune Candida behavior in the gut.


Asunto(s)
Candida albicans/patogenicidad , Interleucina-9/metabolismo , Intestinos/microbiología , Intestinos/patología , Mastocitos/metabolismo , Inmunidad Adaptativa , Animales , Candidiasis/inmunología , Candidiasis/microbiología , Candidiasis/patología , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/patología , Permeabilidad de la Membrana Celular , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Inmunidad Innata , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones Endogámicos C57BL , Receptores de Interleucina-9/metabolismo , Transducción de Señal , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA