Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Front Immunol ; 15: 1369238, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38585273

RESUMEN

Introduction: Exosome-enriched small extracellular vesicles (sEVs) are nanosized organelles known to participate in long distance communication between cells, including in the skin. Atopic dermatitis (AD) is a chronic inflammatory skin disease for which filaggrin (FLG) gene mutations are the strongest genetic risk factor. Filaggrin insufficiency affects multiple cellular function, but it is unclear if sEV-mediated cellular communication originating from the affected keratinocytes is also altered, and if this influences peptide and lipid antigen presentation to T cells in the skin. Methods: Available mRNA and protein expression datasets from filaggrin-insufficient keratinocytes (shFLG), organotypic models and AD skin were used for gene ontology analysis with FunRich tool. sEVs secreted by shFLG and control shC cells were isolated from conditioned media by differential centrifugation. Mass spectrometry was carried out for lipidomic and proteomic profiling of the cells and sEVs. T cell responses to protein, peptide, CD1a lipid antigens, as well as phospholipase A2-digested or intact sEVs were measured by ELISpot and ELISA. Results: Data analysis revealed extensive remodeling of the sEV compartment in filaggrin insufficient keratinocytes, 3D models and the AD skin. Lipidomic profiles of shFLGsEV showed a reduction in the long chain (LCFAs) and polyunsaturated fatty acids (PUFAs; permissive CD1a ligands) and increased content of the bulky headgroup sphingolipids (non-permissive ligands). This resulted in a reduction of CD1a-mediated interferon-γ T cell responses to the lipids liberated from shFLG-generated sEVs in comparison to those induced by sEVs from control cells, and an increase in interleukin 13 secretion. The altered sEV lipidome reflected a generalized alteration in the cellular lipidome in filaggrin-insufficient cells and the skin of AD patients, resulting from a downregulation of key enzymes implicated in fatty acid elongation and desaturation, i.e., enzymes of the ACSL, ELOVL and FADS family. Discussion: We determined that sEVs constitute a source of antigens suitable for CD1a-mediated presentation to T cells. Lipids enclosed within the sEVs secreted on the background of filaggrin insufficiency contribute to allergic inflammation by reducing type 1 responses and inducing a type 2 bias from CD1a-restricted T cells, thus likely perpetuating allergic inflammation in the skin.


Asunto(s)
Dermatitis Atópica , Vesículas Extracelulares , Humanos , Vesículas Extracelulares/metabolismo , Proteínas Filagrina , Inflamación , Proteínas de Filamentos Intermediarios/genética , Queratinocitos , Lípidos , Péptidos/metabolismo , Proteómica , Linfocitos T/metabolismo
2.
Clin Exp Allergy ; 51(11): 1421-1428, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34608691

RESUMEN

BACKGROUND: Palmar hyperlinearity is a feature of ichthyosis vulgaris, the monogenic skin disorder caused by FLG loss-of-function mutations. OBJECTIVE: To investigate how well the presence or absence of hyperlinear palms (HLP) detect FLG genotype in children. METHODS: STARD criteria are used to report this diagnostic accuracy study. Phenotype and genotype data (four most prevalent FLG null mutations) were obtained from a total of 3656 children in three studies: the UK CLOTHES trial (children 1-5 years with moderate-severe atopic eczema); UK BEEP trial (2 year olds at high risk of developing atopic eczema); UK-Irish eczema case collection (0-16 year olds with atopic eczema). All participants included in analyses of HLP as the index test and FLG genotype as the reference were of white European ancestry. RESULTS: Thirty-two percent of participants (1159/3656) had FLG null mutation(s) and 37% (1347/3656) had HLP. In 13% (464/3656), HLP was recorded as 'unsure' or not recorded. The sensitivity and specificity of HLP for detecting FLG mutations in each of the studies was: 67% (95% CI 55-78%) and 75% (67-82%) in CLOTHES; 46% (36-55%) and 89% (86-91%) in BEEP; 72% (68-75%) and 60% (57-62%) in the UK-Irish case collection. Positive and negative likelihood ratios were: 2.73 (1.95-3.81) and 0.44 (0.31-0.62) in CLOTHES; 4.02 (2.99-5.40) and 0.61 (0.52-0.73) in BEEP; 1.79 (1.66-1.93) and 0.47 (0.42-0.53) in the UK-Irish collection. DISCUSSION: Trained observers were able to define palmar hyperlinearity in the majority (3191/3656, 87%) of cases. The presence of HLP is not a reliable sign to detect FLG mutations, but the absence of HLP excludes FLG null genotype with a reasonable degree of certainty.


Asunto(s)
Pruebas Diagnósticas de Rutina , Proteínas Filagrina , Adolescente , Niño , Preescolar , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Lactante , Proteínas de Filamentos Intermediarios/genética , Mutación
3.
J Allergy Clin Immunol ; 144(2): 470-481, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31158401

RESUMEN

BACKGROUND: Atopic dermatitis (AD) is a common, complex, and highly heritable inflammatory skin disease. Genome-wide association studies offer opportunities to identify molecular targets for drug development. A risk locus on chromosome 11q13.5 lies between 2 candidate genes, EMSY and LRRC32 (leucine-rich repeat-containing 32) but the functional mechanisms affecting risk of AD remain unclear. OBJECTIVES: We sought to apply a combination of genomic and molecular analytic techniques to investigate which genes are responsible for genetic risk at this locus and to define mechanisms contributing to atopic skin disease. METHODS: We used interrogation of available genomic and chromosome conformation data in keratinocytes, small interfering RNA (siRNA)-mediated knockdown in skin organotypic culture and functional assessment of barrier parameters, mass spectrometric global proteomic analysis and quantitative lipid analysis, electron microscopy of organotypic skin, and immunohistochemistry of human skin samples. RESULTS: Genomic data indicate active promoters in the genome-wide association study locus and upstream of EMSY; EMSY, LRRC32, and intergenic variants all appear to be within a single topologically associating domain. siRNA-knockdown of EMSY in organotypic culture leads to enhanced development of barrier function, reflecting increased expression of structural and functional proteins, including filaggrin and filaggrin-2, as well as long-chain ceramides. Conversely, overexpression of EMSY in keratinocytes leads to a reduction in markers of barrier formation. Skin biopsy samples from patients with AD show greater EMSY staining in the nucleus, which is consistent with an increased functional effect of this transcriptional control protein. CONCLUSION: Our findings demonstrate an important role for EMSY in transcriptional regulation and skin barrier formation, supporting EMSY inhibition as a therapeutic approach.


Asunto(s)
Dermatitis Atópica/inmunología , Regulación de la Expresión Génica/inmunología , Proteínas de Neoplasias/inmunología , Proteínas Nucleares/inmunología , Proteínas Represoras/inmunología , Piel/inmunología , Transcripción Genética/inmunología , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 11/inmunología , Dermatitis Atópica/genética , Dermatitis Atópica/patología , Femenino , Proteínas Filagrina , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Piel/patología
4.
J Invest Dermatol ; 138(2): 336-343, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28951239

RESUMEN

Staphylococcus aureus is an opportunistic pathogen and variable component of the human microbiota. A characteristic of atopic eczema (AE) is colonization by S. aureus, with exacerbations associated with an increased bacterial burden of the organism. Despite this, the origins and genetic diversity of S. aureus colonizing individual patients during AE disease flares is poorly understood. To examine the microevolution of S. aureus colonization, we deep sequenced S. aureus populations from nine children with moderate to severe AE and 18 non-atopic children asymptomatically carrying S. aureus nasally. Colonization by clonal S. aureus populations was observed in both AE patients and control participants, with all but one of the individuals carrying colonies belonging to a single sequence type. Phylogenetic analysis showed that disease flares were associated with the clonal expansion of the S. aureus population, occurring over a period of weeks to months. There was a significant difference in the genetic backgrounds of S. aureus colonizing AE cases versus controls (Fisher exact test, P = 0.03). Examination of intra-host genetic heterogeneity of the colonizing S. aureus populations identified evidence of within-host selection in the AE patients, with AE variants being potentially selectively advantageous for intracellular persistence and treatment resistance.


Asunto(s)
Antibacterianos/farmacología , Dermatitis Atópica/microbiología , Farmacorresistencia Bacteriana/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Antibacterianos/uso terapéutico , Estudios de Casos y Controles , Niño , Preescolar , Evolución Clonal/efectos de los fármacos , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/patología , Femenino , Interacciones Microbiota-Huesped/genética , Humanos , Lactante , Masculino , Filogenia , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/patología , Staphylococcus aureus/aislamiento & purificación , Brote de los Síntomas
5.
J Allergy Clin Immunol ; 139(4): 1228-1241, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27913303

RESUMEN

BACKGROUND: Filaggrin, which is encoded by the filaggrin gene (FLG), is an important component of the skin's barrier to the external environment, and genetic defects in FLG strongly associate with atopic dermatitis (AD). However, not all patients with AD have FLG mutations. OBJECTIVE: We hypothesized that these patients might possess other defects in filaggrin expression and processing contributing to barrier disruption and AD, and therefore we present novel therapeutic targets for this disease. RESULTS: We describe the relationship between the mechanistic target of rapamycin complex 1/2 protein subunit regulatory associated protein of the MTOR complex 1 (RAPTOR), the serine/threonine kinase V-Akt murine thymoma viral oncogene homolog 1 (AKT1), and the protease cathepsin H (CTSH), for which we establish a role in filaggrin expression and processing. Increased RAPTOR levels correlated with decreased filaggrin expression in patients with AD. In keratinocyte cell cultures RAPTOR upregulation or AKT1 short hairpin RNA knockdown reduced expression of the protease CTSH. Skin of CTSH-deficient mice and CTSH short hairpin RNA knockdown keratinocytes showed reduced filaggrin processing, and the mouse had both impaired skin barrier function and a mild proinflammatory phenotype. CONCLUSION: Our findings highlight a novel and potentially treatable signaling axis controlling filaggrin expression and processing that is defective in patients with AD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Catepsina H/metabolismo , Dermatitis Atópica/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Western Blotting , Catepsina H/deficiencia , Dermatitis Atópica/patología , Proteínas Filagrina , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Queratinocitos/metabolismo , Queratinocitos/patología , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Reguladora Asociada a mTOR , Piel/metabolismo , Piel/patología
6.
J Allergy Clin Immunol ; 134(1): 82-91, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24880632

RESUMEN

BACKGROUND: Atopic dermatitis (AD; eczema) is characterized by a widespread abnormality in cutaneous barrier function and propensity to inflammation. Filaggrin is a multifunctional protein and plays a key role in skin barrier formation. Loss-of-function mutations in the gene encoding filaggrin (FLG) are a highly significant risk factor for atopic disease, but the molecular mechanisms leading to dermatitis remain unclear. OBJECTIVE: We sought to interrogate tissue-specific variations in the expressed genome in the skin of children with AD and to investigate underlying pathomechanisms in atopic skin. METHODS: We applied single-molecule direct RNA sequencing to analyze the whole transcriptome using minimal tissue samples. Uninvolved skin biopsy specimens from 26 pediatric patients with AD were compared with site-matched samples from 10 nonatopic teenage control subjects. Cases and control subjects were screened for FLG genotype to stratify the data set. RESULTS: Two thousand four hundred thirty differentially expressed genes (false discovery rate, P < .05) were identified, of which 211 were significantly upregulated and 490 downregulated by greater than 2-fold. Gene ontology terms for "extracellular space" and "defense response" were enriched, whereas "lipid metabolic processes" were downregulated. The subset of FLG wild-type cases showed dysregulation of genes involved with lipid metabolism, whereas filaggrin haploinsufficiency affected global gene expression and was characterized by a type 1 interferon-mediated stress response. CONCLUSION: These analyses demonstrate the importance of extracellular space and lipid metabolism in atopic skin pathology independent of FLG genotype, whereas an aberrant defense response is seen in subjects with FLG mutations. Genotype stratification of the large data set has facilitated functional interpretation and might guide future therapy development.


Asunto(s)
Dermatitis Atópica/genética , Proteínas de Filamentos Intermediarios/genética , Piel/metabolismo , Transcripción Genética/inmunología , Adolescente , Estudios de Casos y Controles , Niño , Dermatitis Atópica/inmunología , Dermatitis Atópica/metabolismo , Dermatitis Atópica/patología , Espacio Extracelular/inmunología , Femenino , Proteínas Filagrina , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Proteínas de Filamentos Intermediarios/inmunología , Metabolismo de los Lípidos/inmunología , Masculino , Piel/inmunología , Piel/patología , Adulto Joven
8.
Nat Genet ; 44(11): 1272-6, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23064416

RESUMEN

Palmoplantar keratodermas (PPKs) are a group of disorders that are diagnostically and therapeutically problematic in dermatogenetics. Punctate PPKs are characterized by circumscribed hyperkeratotic lesions on the palms and soles with considerable heterogeneity. In 18 families with autosomal dominant punctate PPK, we report heterozygous loss-of-function mutations in AAGAB, encoding α- and γ-adaptin-binding protein p34, located at a previously linked locus at 15q22. α- and γ-adaptin-binding protein p34, a cytosolic protein with a Rab-like GTPase domain, was shown to bind both clathrin adaptor protein complexes, indicating a role in membrane trafficking. Ultrastructurally, lesional epidermis showed abnormalities in intracellular vesicle biology. Immunohistochemistry showed hyperproliferation within the punctate lesions. Knockdown of AAGAB in keratinocytes led to increased cell division, which was linked to greatly elevated epidermal growth factor receptor (EGFR) protein expression and tyrosine phosphorylation. We hypothesize that p34 deficiency may impair endocytic recycling of growth factor receptors such as EGFR, leading to increased signaling and cellular proliferation.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular , Proteínas Portadoras/genética , Haploinsuficiencia , Poroqueratosis/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Mapeo Cromosómico , Citosol/ultraestructura , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Linaje , Poroqueratosis/metabolismo , Unión Proteica , Proteínas/genética , Proteínas/metabolismo
9.
Semin Cutan Med Surg ; 27(2): 128-37, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18620134

RESUMEN

The discovery that null mutations in the filaggrin gene (FLG) are associated with atopic eczema represents the single most significant breakthrough in understanding the genetic basis of this complex disorder. The association has been replicated in multiple independent studies during the past 2 years with the use of various methodologies, from populations in Europe, the United States, and Japan. Filaggrin plays a key role in epidermal barrier function, and its association with atopic eczema emphasizes the importance of barrier dysfunction in eczema pathogenesis. This review aims to summarize the current state of knowledge regarding the role of FLG mutations in ichthyosis vulgaris, atopic eczema, and other skin disorders, with an emphasis on potential clinical applications. Further research is needed to clarify the precise role of filaggrin in skin and systemic atopic disease, to pave the way for novel therapeutic interventions.


Asunto(s)
ADN/genética , Dermatitis Atópica/genética , Proteínas de Filamentos Intermediarios/genética , Mutación , Proteínas Filagrina , Predisposición Genética a la Enfermedad , Humanos , Fosfoproteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA