Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Genet Med ; 25(4): 100018, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36681873

RESUMEN

PURPOSE: Within the Solve-RD project (https://solve-rd.eu/), the European Reference Network for Intellectual disability, TeleHealth, Autism and Congenital Anomalies aimed to investigate whether a reanalysis of exomes from unsolved cases based on ClinVar annotations could establish additional diagnoses. We present the results of the "ClinVar low-hanging fruit" reanalysis, reasons for the failure of previous analyses, and lessons learned. METHODS: Data from the first 3576 exomes (1522 probands and 2054 relatives) collected from European Reference Network for Intellectual disability, TeleHealth, Autism and Congenital Anomalies was reanalyzed by the Solve-RD consortium by evaluating for the presence of single-nucleotide variant, and small insertions and deletions already reported as (likely) pathogenic in ClinVar. Variants were filtered according to frequency, genotype, and mode of inheritance and reinterpreted. RESULTS: We identified causal variants in 59 cases (3.9%), 50 of them also raised by other approaches and 9 leading to new diagnoses, highlighting interpretation challenges: variants in genes not known to be involved in human disease at the time of the first analysis, misleading genotypes, or variants undetected by local pipelines (variants in off-target regions, low quality filters, low allelic balance, or high frequency). CONCLUSION: The "ClinVar low-hanging fruit" analysis represents an effective, fast, and easy approach to recover causal variants from exome sequencing data, herewith contributing to the reduction of the diagnostic deadlock.


Asunto(s)
Discapacidad Intelectual , Humanos , Secuenciación del Exoma , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Alelos , Genotipo
2.
PLoS Pathog ; 10(9): e1004351, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25187968

RESUMEN

NOD1 is an intracellular pathogen recognition receptor that contributes to anti-bacterial innate immune responses, adaptive immunity and tissue homeostasis. NOD1-induced signaling relies on actin remodeling, however, the details of the connection of NOD1 and the actin cytoskeleton remained elusive. Here, we identified in a druggable-genome wide siRNA screen the cofilin phosphatase SSH1 as a specific and essential component of the NOD1 pathway. We show that depletion of SSH1 impaired pathogen induced NOD1 signaling evident from diminished NF-κB activation and cytokine release. Chemical inhibition of actin polymerization using cytochalasin D rescued the loss of SSH1. We further demonstrate that NOD1 directly interacted with SSH1 at F-actin rich sites. Finally, we show that enhanced cofilin activity is intimately linked to NOD1 signaling. Our data thus provide evidence that NOD1 requires the SSH1/cofilin network for signaling and to detect bacterial induced changes in actin dynamics leading to NF-κB activation and innate immune responses.


Asunto(s)
Actinas/metabolismo , Cofilina 1/metabolismo , Disentería Bacilar/microbiología , Proteína Adaptadora de Señalización NOD1/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Shigella flexneri/fisiología , Actinas/química , Western Blotting , Células Cultivadas , Cofilina 1/genética , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Regulación de la Expresión Génica , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Inflamación , Mediadores de Inflamación/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/antagonistas & inhibidores , Proteína Adaptadora de Señalización NOD1/genética , Fosfoproteínas Fosfatasas/genética , Fosforilación , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
3.
J Biol Chem ; 289(33): 22900-22914, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24958724

RESUMEN

Following activation, the cytoplasmic pattern recognition receptor nucleotide-binding oligomerization domain-containing protein 1 (NOD1) interacts with its adaptor protein receptor-interacting protein 2 (RIP2) to propagate immune signaling and initiate a proinflammatory immune response. This interaction is mediated by the caspase recruitment domain (CARD) of both proteins. Polymorphisms in immune proteins can affect receptor function and predispose individuals to specific autoinflammatory disorders. In this report, we show that mutations in helix 2 of the CARD of NOD1 disrupted receptor function but did not interfere with RIP2 interaction. In particular, N43S, a rare polymorphism, resulted in receptor dysfunction despite retaining normal cellular localization, protein folding, and an ability to interact with RIP2. Mutation of Asn-43 resulted in an increased tendency to form dimers, which we propose is the source of this dysfunction. We also demonstrate that mutation of Lys-443 and Tyr-474 in RIP2 disrupted the interaction with NOD1. Mapping the key residues involved in the interaction between NOD1 and RIP2 to the known structures of CARD complexes revealed the likely involvement of both type I and type III interfaces in the NOD1·RIP2 complex. Overall we demonstrate that the NOD1-RIP2 signaling axis is more complex than previously assumed, that simple engagement of RIP2 is insufficient to mediate signaling, and that the interaction between NOD1 and RIP2 constitutes multiple CARD-CARD interfaces.


Asunto(s)
Proteína Adaptadora de Señalización NOD1/metabolismo , Multimerización de Proteína/fisiología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Transducción de Señal/fisiología , Células HEK293 , Humanos , Mutación , Proteína Adaptadora de Señalización NOD1/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética
4.
Eur J Hum Genet ; 29(9): 1325-1331, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34075208

RESUMEN

For the first time in Europe hundreds of rare disease (RD) experts team up to actively share and jointly analyse existing patient's data. Solve-RD is a Horizon 2020-supported EU flagship project bringing together >300 clinicians, scientists, and patient representatives of 51 sites from 15 countries. Solve-RD is built upon a core group of four European Reference Networks (ERNs; ERN-ITHACA, ERN-RND, ERN-Euro NMD, ERN-GENTURIS) which annually see more than 270,000 RD patients with respective pathologies. The main ambition is to solve unsolved rare diseases for which a molecular cause is not yet known. This is achieved through an innovative clinical research environment that introduces novel ways to organise expertise and data. Two major approaches are being pursued (i) massive data re-analysis of >19,000 unsolved rare disease patients and (ii) novel combined -omics approaches. The minimum requirement to be eligible for the analysis activities is an inconclusive exome that can be shared with controlled access. The first preliminary data re-analysis has already diagnosed 255 cases form 8393 exomes/genome datasets. This unprecedented degree of collaboration focused on sharing of data and expertise shall identify many new disease genes and enable diagnosis of many so far undiagnosed patients from all over Europe.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Difusión de la Información , Colaboración Intersectorial , Enfermedades Raras/genética , Conferencias de Consenso como Asunto , Europa (Continente) , Enfermedades Genéticas Congénitas/diagnóstico , Pruebas Genéticas/métodos , Humanos , Enfermedades Raras/diagnóstico , Secuenciación del Exoma/métodos
5.
Orphanet J Rare Dis ; 15(1): 206, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32787960

RESUMEN

BACKGROUND: Rare diseases are individually rare but globally affect around 6% of the population, and in over 70% of cases are genetically determined. Their rarity translates into a delayed diagnosis, with 25% of patients waiting 5 to 30 years for one. It is essential to raise awareness of patients and clinicians of existing gene and variant-specific therapeutics at the time of diagnosis to avoid that treatment delays add up to the diagnostic odyssey of rare diseases' patients and their families. AIMS: This paper aims to provide guidance and give detailed instructions on how to write homogeneous systematic reviews of rare diseases' treatments in a manner that allows the capture of the results in a computer-accessible form. The published results need to comply with the FAIR guiding principles for scientific data management and stewardship to facilitate the extraction of datasets that are easily transposable into machine-actionable information. The ultimate purpose is the creation of a database of rare disease treatments ("Treatabolome") at gene and variant levels as part of the H2020 research project Solve-RD. RESULTS: Each systematic review follows a written protocol to address one or more rare diseases in which the authors are experts. The bibliographic search strategy requires detailed documentation to allow its replication. Data capture forms should be built to facilitate the filling of a data capture spreadsheet and to record the application of the inclusion and exclusion criteria to each search result. A PRISMA flowchart is required to provide an overview of the processes of search and selection of papers. A separate table condenses the data collected during the Systematic Review, appraised according to their level of evidence. CONCLUSIONS: This paper provides a template that includes the instructions for writing FAIR-compliant systematic reviews of rare diseases' treatments that enables the assembly of a Treatabolome database that complement existing diagnostic and management support tools with treatment awareness data.


Asunto(s)
Manejo de Datos , Enfermedades Raras , Humanos , Enfermedades Raras/genética , Enfermedades Raras/terapia , Proyectos de Investigación , Revisiones Sistemáticas como Asunto , Escritura
10.
Innate Immun ; 18(1): 100-11, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21310790

RESUMEN

Nucleotide-binding oligomerization domain-containing protein (NOD)1 and NOD2 are intracellular pattern recognition receptors (PRRs) of the nucleotide-binding domain and leucine-rich repeat containing (NLR) gene family involved in innate immune responses. Their centrally located NACHT domain displays ATPase activity and is necessary for activation and oligomerization leading to inflammatory signaling responses. Mutations affecting key residues of the ATPase domain of NOD2 are linked to severe auto-inflammatory diseases, such as Blau syndrome and early-onset sarcoidosis. By mutational dissection of the ATPase domain function, we show that the NLR-specific extended Walker B box (DGhDE) can functionally replace the canonical Walker B sequence (DDhWD) found in other ATPases. A requirement for an intact Walker A box and the magnesium-co-ordinating aspartate of the classical Walker B box suggest that an initial ATP hydrolysis step is necessary for activation of both NOD1 and NOD2. In contrast, a Blau-syndrome associated mutation located in the extended Walker B box of NOD2 that results in higher autoactivation and ligand-induced signaling does not affect NOD1 function. Moreover, mutation of a conserved histidine in the NACHT domain also has contrasting effects on NOD1 and NOD2 mediated NF-κB activation. We conclude that these two NLRs employ different modes of activation and propose distinct models for activation of NOD1 and NOD2.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Enfermedades de los Nervios Craneales/genética , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD2/genética , Sarcoidosis/genética , Sinovitis/genética , Uveítis/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Artritis , Enfermedades de los Nervios Craneales/inmunología , Análisis Mutacional de ADN , Células HeLa , Humanos , Inmunidad Innata , Mutación/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas NLR , Proteína Adaptadora de Señalización NOD1/inmunología , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/inmunología , Proteína Adaptadora de Señalización NOD2/metabolismo , Polimorfismo Genético , Estructura Terciaria de Proteína/genética , Sarcoidosis/inmunología , Transducción de Señal/genética , Sinovitis/inmunología , Activación Transcripcional/genética , Uveítis/inmunología
11.
PLoS One ; 7(7): e41255, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22829933

RESUMEN

NOD2, the nucleotide-binding domain and leucine-rich repeat containing gene family (NLR) member 2 is involved in mediating antimicrobial responses. Dysfunctional NOD2 activity can lead to severe inflammatory disorders, but the regulation of NOD2 is still poorly understood. Recently, proteins of the tripartite motif (TRIM) protein family have emerged as regulators of innate immune responses by acting as E3 ubiquitin ligases. We identified TRIM27 as a new specific binding partner for NOD2. We show that NOD2 physically interacts with TRIM27 via the nucleotide-binding domain, and that NOD2 activation enhances this interaction. Dependent on functional TRIM27, ectopically expressed NOD2 is ubiquitinated with K48-linked ubiquitin chains followed by proteasomal degradation. Accordingly, TRIM27 affects NOD2-mediated pro-inflammatory responses. NOD2 mutations are linked to susceptibility to Crohn's disease. We found that TRIM27 expression is increased in Crohn's disease patients, underscoring a physiological role of TRIM27 in regulating NOD2 signaling. In HeLa cells, TRIM27 is partially localized in the nucleus. We revealed that ectopically expressed NOD2 can shuttle to the nucleus in a Walker A dependent manner, suggesting that NOD2 and TRIM27 might functionally cooperate in the nucleus.We conclude that TRIM27 negatively regulates NOD2-mediated signaling by degradation of NOD2 and suggest that TRIM27 could be a new target for therapeutic intervention in NOD2-associated diseases.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Línea Celular , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Técnicas In Vitro , Microscopía Fluorescente , Proteína Adaptadora de Señalización NOD2/genética , Proteínas Nucleares/genética , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitinación/genética , Ubiquitinación/fisiología
12.
Methods Mol Biol ; 748: 107-19, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21701969

RESUMEN

Nod1 and Nod2 are pattern recognition receptors of the mammalian innate immune system. They respond to bacterial peptidoglycan fragments and are implicated in host defense against a variety of -different bacterial pathogens. Recent studies furthermore support additional functions of these proteins in the control of adaptive immune responses and intestinal homeostasis. Activation of Nod1 and Nod2 by their cognate elicitors triggers inflammatory responses driven by the activation of NF-κB and MAPK pathways. In this chapter, we describe a quick and reliable cell-based assay using a luciferase reporter to measure Nod1- and Nod2-mediated NF-κB activation. The described protocol was successfully applied to analyze the influences of overexpressed proteins and siRNA-mediated knock-down to provide new insights into the regulation of Nod1/2-specific signaling pathways. Furthermore, this method is well suited for downscaling to high-throughput screening applications.


Asunto(s)
Bioensayo/métodos , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Línea Celular , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD2/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
Gut Microbes ; 2(5): 274-9, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22067940

RESUMEN

Outer membrane vesicles (OMVs) are released from many Gram-negative bacteria. OMVs interact with and are taken up by human cells. We and others have now showed that OMVs contain peptidoglycan, which is sensed mainly by the pattern-recognition receptor NOD1 in the cytoplasm of host cells. Vibrio cholerae is clinically important as one of the causative agents of severe dehydrating diarrhea in humans. We showed that non-O1 non-O139 V. cholerae (NOVC) strains of V. cholera produce OMVs. Of note, we revealed that NOVC can evade NOD1-mediated immune surveillance by the quorum sensing machinery. Here we review these recent findings and discuss the relevance for our understanding of bacterial infections and innate immune responses.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Cólera/inmunología , Percepción de Quorum , Vesículas Secretoras/metabolismo , Vibrio cholerae/fisiología , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Cólera/genética , Cólera/microbiología , Humanos , Inmunidad Innata , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD1/inmunología , Vesículas Secretoras/genética , Vibrio cholerae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA