Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Mol Metab ; 84: 101938, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631478

RESUMEN

OBJECTIVE: The peroxisome proliferator-activated receptor α (PPARα) is a transcription factor driving target genes involved in fatty acid ß-oxidation. To what extent various PPARα interacting proteins may assist its function as a transcription factor is incompletely understood. An ORFeome-wide unbiased mammalian protein-protein interaction trap (MAPPIT) using PPARα as bait revealed a PPARα-ligand-dependent interaction with the orphan nuclear receptor estrogen-related receptor α (ERRα). The goal of this study was to characterize the nature of the interaction in depth and to explore whether it was of physiological relevance. METHODS: We used orthogonal protein-protein interaction assays and pharmacological inhibitors of ERRα in various systems to confirm a functional interaction and study the impact of crosstalk mechanisms. To characterize the interaction surfaces and contact points we applied a random mutagenesis screen and structural overlays. We pinpointed the extent of reciprocal ligand effects of both nuclear receptors via coregulator peptide recruitment assays. On PPARα targets revealed from a genome-wide transcriptome analysis, we performed an ERRα chromatin immunoprecipitation analysis on both fast and fed mouse livers. RESULTS: Random mutagenesis scanning of PPARα's ligand-binding domain and coregulator profiling experiments supported the involvement of (a) bridging coregulator(s), while recapitulation of the interaction in vitro indicated the possibility of a trimeric interaction with RXRα. The PPARα·ERRα interaction depends on 3 C-terminal residues within helix 12 of ERRα and is strengthened by both PGC1α and serum deprivation. Pharmacological inhibition of ERRα decreased the interaction of ERRα to ligand-activated PPARα and revealed a transcriptome in line with enhanced mRNA expression of prototypical PPARα target genes, suggesting a role for ERRα as a transcriptional repressor. Strikingly, on other PPARα targets, including the isolated PDK4 enhancer, ERRα behaved oppositely. Chromatin immunoprecipitation analyses demonstrate a PPARα ligand-dependent ERRα recruitment onto chromatin at PPARα-binding regions, which is lost following ERRα inhibition in fed mouse livers. CONCLUSIONS: Our data support the coexistence of multiple layers of transcriptional crosstalk mechanisms between PPARα and ERRα, which may serve to finetune the activity of PPARα as a nutrient-sensing transcription factor.


Asunto(s)
Receptor Relacionado con Estrógeno ERRalfa , PPAR alfa , Receptores de Estrógenos , PPAR alfa/metabolismo , PPAR alfa/genética , Animales , Ratones , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Humanos , Regulación de la Expresión Génica , Células HEK293 , Masculino , Ratones Endogámicos C57BL , Unión Proteica , Hígado/metabolismo
2.
Cell Rep Methods ; 3(11): 100641, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37963462

RESUMEN

Protein mutagenesis is essential for unveiling the molecular mechanisms underlying protein function in health, disease, and evolution. In the past decade, deep mutational scanning methods have evolved to support the functional analysis of nearly all possible single-amino acid changes in a protein of interest. While historically these methods were developed in lower organisms such as E. coli and yeast, recent technological advancements have resulted in the increased use of mammalian cells, particularly for studying proteins involved in human disease. These advancements will aid significantly in the classification and interpretation of variants of unknown significance, which are being discovered at large scale due to the current surge in the use of whole-genome sequencing in clinical contexts. Here, we explore the experimental aspects of deep mutational scanning studies in mammalian cells and report the different methods used in each step of the workflow, ultimately providing a useful guide toward the design of such studies.


Asunto(s)
Escherichia coli , Proteínas , Animales , Humanos , Mutación , Proteínas/genética , Mutagénesis , Aminoácidos , Mamíferos/genética
3.
Cells ; 12(12)2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37371105

RESUMEN

The glucocorticoid receptor α (GRα) is a member of the nuclear receptor superfamily and functions as a glucocorticoid (GC)-responsive transcription factor. GR can halt inflammation and kill off cancer cells, thus explaining the widespread use of glucocorticoids in the clinic. However, side effects and therapy resistance limit GR's therapeutic potential, emphasizing the importance of resolving all of GR's context-specific action mechanisms. Fortunately, the understanding of GR structure, conformation, and stoichiometry in the different GR-controlled biological pathways is now gradually increasing. This information will be crucial to close knowledge gaps on GR function. In this review, we focus on the various domains and mechanisms of action of GR, all from a structural perspective.


Asunto(s)
Receptores de Glucocorticoides , Humanos , Glucocorticoides/farmacología , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción
4.
J Immunother Cancer ; 9(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34772757

RESUMEN

BACKGROUND: Clinical success of therapeutic cancer vaccines depends on the ability to mount strong and durable antitumor T cell responses. To achieve this, potent cellular adjuvants are highly needed. Interleukin-1ß (IL-1ß) acts on CD8+ T cells and promotes their expansion and effector differentiation, but toxicity and undesired tumor-promoting side effects hamper efficient clinical application of this cytokine. METHODS: This 'cytokine problem' can be solved by use of AcTakines (Activity-on-Target cytokines), which represent fusions between low-activity cytokine mutants and cell type-specific single-domain antibodies. AcTakines deliver cytokine activity to a priori selected cell types and as such evade toxicity and unwanted off-target side effects. Here, we employ subcutaneous melanoma and lung carcinoma models to evaluate the antitumor effects of AcTakines. RESULTS: In this work, we use an IL-1ß-based AcTakine to drive proliferation and effector functionality of antitumor CD8+ T cells without inducing measurable toxicity. AcTakine treatment enhances diversity of the T cell receptor repertoire and empowers adoptive T cell transfer. Combination treatment with a neovasculature-targeted tumor necrosis factor (TNF) AcTakine mediates full tumor eradication and establishes immunological memory that protects against secondary tumor challenge. Interferon-γ was found to empower this AcTakine synergy by sensitizing the tumor microenvironment to TNF. CONCLUSIONS: Our data illustrate that anticancer cellular immunity can be safely promoted with an IL-1ß-based AcTakine, which synergizes with other immunotherapies for efficient tumor destruction.


Asunto(s)
Inmunoterapia/métodos , Interleucina-1/metabolismo , Neoplasias/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones
5.
Sci Transl Med ; 13(621): eabi7826, 2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34609205

RESUMEN

Broadly neutralizing antibodies are an important treatment for individuals with coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Antibody-based therapeutics are also essential for pandemic preparedness against future Sarbecovirus outbreaks. Camelid-derived single domain antibodies (VHHs) exhibit potent antimicrobial activity and are being developed as SARS-CoV-2­neutralizing antibody-like therapeutics. Here, we identified VHHs that neutralize both SARS-CoV-1 and SARS-CoV-2, including now circulating variants. We observed that the VHHs bound to a highly conserved epitope in the receptor binding domain of the viral spike protein that is difficult to access for human antibodies. Structure-guided molecular modeling, combined with rapid yeast-based prototyping, resulted in an affinity enhanced VHH-human immunoglobulin G1 Fc fusion molecule with subnanomolar neutralizing activity. This VHH-Fc fusion protein, produced in and purified from cultured Chinese hamster ovary cells, controlled SARS-CoV-2 replication in prophylactic and therapeutic settings in mice expressing human angiotensin converting enzyme 2 and in hamsters infected with SARS-CoV-2. These data led to affinity-enhanced selection of the VHH, XVR011, a stable anti­COVID-19 biologic that is now being evaluated in the clinic.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Humanos , Modelos Animales , SARS-CoV-2
6.
NPJ Vaccines ; 5(1): 64, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32714571

RESUMEN

Annual administration and reformulation of influenza vaccines is required for protection against seasonal infections. However, the induction of strong and long-lasting T cells is critical to reach broad and potentially lifelong antiviral immunity. The NLRP3 inflammasome and its product interleukin-1ß (IL-1ß) are pivotal mediators of cellular immune responses to influenza, yet, overactivation of these systems leads to side effects, which hamper clinical applications. Here, we present a bypass around these toxicities by targeting the activity of IL-1ß to CD8+ T cells. Using this approach, we demonstrate safe inclusion of IL-1ß as an adjuvant in vaccination strategies, leading to full protection of mice against a high influenza virus challenge dose by raising potent T cell responses. In conclusion, this paper proposes a class of IL-1ß-based vaccine adjuvants and also provides further insight in the mechanics of cellular immune responses driven by IL-1ß.

7.
EMBO Mol Med ; 12(2): e11223, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31912630

RESUMEN

Systemic toxicities have severely limited the clinical application of tumor necrosis factor (TNF) as an anticancer agent. Activity-on-Target cytokines (AcTakines) are a novel class of immunocytokines with improved therapeutic index. A TNF-based AcTakine targeted to CD13 enables selective activation of the tumor neovasculature without any detectable toxicity in vivo. Upregulation of adhesion markers supports enhanced T-cell infiltration leading to control or elimination of solid tumors by, respectively, CAR T cells or a combination therapy with CD8-targeted type I interferon AcTakine. Co-treatment with a CD13-targeted type II interferon AcTakine leads to very rapid destruction of the tumor neovasculature and complete regression of large, established tumors. As no tumor markers are needed, safe and efficacious elimination of a broad range of tumor types becomes feasible.


Asunto(s)
Inmunoterapia , Neoplasias , Factor de Necrosis Tumoral alfa , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/terapia
8.
Hum Mutat ; 41(5): 998-1011, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31999394

RESUMEN

Inactivating variants in the centrosomal CEP78 gene have been found in cone-rod dystrophy with hearing loss (CRDHL), a particular phenotype distinct from Usher syndrome. Here, we identified and functionally characterized the first CEP78 missense variant c.449T>C, p.(Leu150Ser) in three CRDHL families. The variant was found in a biallelic state in two Belgian families and in a compound heterozygous state-in trans with c.1462-1G>T-in a third German family. Haplotype reconstruction showed a founder effect. Homology modeling revealed a detrimental effect of p.(Leu150Ser) on protein stability, which was corroborated in patients' fibroblasts. Elongated primary cilia without clear ultrastructural abnormalities in sperm or nasal brushes suggest impaired cilia assembly. Two affected males from different families displayed sperm abnormalities causing infertility. One of these is a heterozygous carrier of a complex allele in SPAG17, a ciliary gene previously associated with autosomal recessive male infertility. Taken together, our data indicate that a missense founder allele in CEP78 underlies the same sensorineural CRDHL phenotype previously associated with inactivating variants. Interestingly, the CEP78 phenotype has been possibly expanded with male infertility. Finally, CEP78 loss-of-function variants may have an underestimated role in misdiagnosed Usher syndrome, with or without sperm abnormalities.


Asunto(s)
Alelos , Proteínas de Ciclo Celular/genética , Distrofias de Conos y Bastones/genética , Efecto Fundador , Pérdida Auditiva/genética , Infertilidad Masculina/genética , Mutación Missense , Adolescente , Proteínas de Ciclo Celular/química , Cilios/metabolismo , Cilios/ultraestructura , Distrofias de Conos y Bastones/diagnóstico , Análisis Mutacional de ADN , Femenino , Fibroblastos/metabolismo , Genotipo , Pérdida Auditiva/diagnóstico , Humanos , Infertilidad Masculina/diagnóstico , Masculino , Persona de Mediana Edad , Modelos Moleculares , Linaje , Fenotipo , Conformación Proteica , Relación Estructura-Actividad , Síndrome , Secuenciación del Exoma
9.
Int J Mol Sci ; 20(9)2019 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-31060328

RESUMEN

The small GTase Arf6 has several important functions in intracellular vesicular trafficking and regulates the recycling of different types of cargo internalized via clathrin-dependent or -independent endocytosis. It activates the lipid modifying enzymes PIP 5-kinase and phospholipase D, promotes actin polymerization, and affects several functionally distinct processes in the cell. Arf6 is used for the phagocytosis of pathogens and can be directly or indirectly targeted by various pathogens to block phagocytosis or induce the uptake of intracellular pathogens. Arf6 is also used in the signaling of Toll-like receptors and in the activation of NADPH oxidases. In this review, we first give an overview of the different roles and mechanisms of action of Arf6 and then focus on its role in innate immunity and host-pathogen interactions.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Factor 6 de Ribosilación del ADP , Animales , Biomarcadores , Activación Enzimática , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , Humanos , Fagocitosis/genética , Fagocitosis/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Virulencia
10.
Int J Mol Sci ; 20(9)2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31027327

RESUMEN

The MAPPIT (mammalian protein protein interaction trap) method allows high-throughput detection of protein interactions by very simple co-transfection of three plasmids in HEK293T cells, followed by a luciferase readout. MAPPIT detects a large percentage of all protein interactions, including those requiring posttranslational modifications and endogenous or exogenous ligands. Here, we present a straightforward method that allows detailed mapping of interaction interfaces via MAPPIT. The method provides insight into the interaction mechanism and reveals how this is affected by disease-associated mutations. By combining error-prone polymerase chain reaction (PCR) for random mutagenesis, 96-well DNA prepping, Sanger sequencing, and MAPPIT via 384-well transfections, we test the effects of a large number of mutations of a selected protein on its protein interactions. The entire screen takes less than three months and interactions with multiple partners can be studied in parallel. The effect of mutations on the MAPPIT readout is mapped on the protein structure, allowing unbiased identification of all putative interaction sites. We have thus far analysed 6 proteins and mapped their interfaces for 16 different interaction partners. Our method is broadly applicable as the required tools are simple and widely available.


Asunto(s)
Mutagénesis/genética , Mapeo de Interacción de Proteínas/métodos , Animales , Humanos , Unión Proteica
11.
Hum Mutat ; 40(5): 539-551, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30668888

RESUMEN

Sorsby fundus dystrophy (SFD) is a macular degeneration caused by mutations in TIMP3, the majority of which introduce a novel cysteine. However, the exact molecular mechanisms underlying SFD remain unknown. We aimed to provide novel insights into the functional consequences of a distinct N-terminal mutation. Haplotype reconstruction in three SFD families revealed that the identified c.113C>G, p.(Ser38Cys) mutation is a founder in Belgian and northern French families with a late-onset SFD phenotype. Functional consequences of the p.(Ser38Cys) mutation were investigated by high-resolution Western blot analysis of wild type and mutant TIMP3 using patient fibroblasts and in vitro generated proteins, and by molecular modeling of TIMP3 and its interaction partners. We could not confirm a previous hypothesis on dimerization of mutant TIMP3 proteins. However, we identified aberrant intramolecular disulfide bonding. Our data provide evidence for disruption of the established Cys36-Cys143 disulfide bond and formation of a novel Cys36-Cys38 bond, possibly associated with increased glycosylation of the protein. In conclusion, we propose a novel pathogenetic mechanism underlying the p.(Ser38Cys) TIMP3 founder mutation involving intramolecular disulfide bonding. These results provide new insights into the pathogenesis of SFD and other retinopathies linked to mutations in TIMP3, such as age-related macular degeneration.


Asunto(s)
Efecto Fundador , Degeneración Macular/diagnóstico , Degeneración Macular/genética , Mutación , Dominios y Motivos de Interacción de Proteínas , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidor Tisular de Metaloproteinasa-3/genética , Anciano , Disulfuros , Femenino , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Haplotipos , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Peso Molecular , Linaje , Conformación Proteica , Relación Estructura-Actividad , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
12.
Genet Med ; 21(4): 1028, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30607024

RESUMEN

The original version of this Article contained an incorrect version of Fig. 3, which included two variants initially shown in black text in Fig. 3a that the authors removed from the final manuscript. The correct version of Fig. 3 without the two variants now appears in the PDF and HTML versions of the Article.

13.
Genet Med ; 21(6): 1319-1329, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30377383

RESUMEN

PURPOSE: RAX2 encodes a homeobox-containing transcription factor, in which four monoallelic pathogenic variants have been described in autosomal dominant cone-dominated retinal disease. METHODS: Exome sequencing in a European cohort with inherited retinal disease (IRD) (n = 2086) was combined with protein structure modeling of RAX2 missense variants, bioinformatics analysis of deletion breakpoints, haplotyping of RAX2 variant c.335dup, and clinical assessment of biallelic RAX2-positive cases and carrier family members. RESULTS: Biallelic RAX2 sequence and structural variants were found in five unrelated European index cases, displaying nonsyndromic autosomal recessive retinitis pigmentosa (ARRP) with an age of onset ranging from childhood to the mid-40s (average mid-30s). Protein structure modeling points to loss of function of the novel recessive missense variants and to a dominant-negative effect of the reported dominant RAX2 alleles. Structural variants were fine-mapped to disentangle their underlying mechanisms. Haplotyping of c.335dup in two cases suggests a common ancestry. CONCLUSION: This study supports a role for RAX2 as a novel disease gene for recessive IRD, broadening the mutation spectrum from sequence to structural variants and revealing a founder effect. The identification of biallelic RAX2 pathogenic variants in five unrelated families shows that RAX2 loss of function may be a nonnegligible cause of IRD in unsolved ARRP cases.


Asunto(s)
Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Retinitis Pigmentosa/genética , Factores de Transcripción/genética , Adulto , Análisis Mutacional de ADN/métodos , Proteínas del Ojo/metabolismo , Proteínas del Ojo/fisiología , Femenino , Genes Recesivos/genética , Estudios de Asociación Genética/métodos , Genotipo , Haplotipos/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/fisiología , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Mutación Missense/genética , Linaje , Fenotipo , Retina/metabolismo , Retina/patología , Enfermedades de la Retina/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Población Blanca/genética
14.
Sci Rep ; 8(1): 6673, 2018 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-29691449

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

16.
Genet Med ; 20(7): 717-727, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29261182

RESUMEN

PURPOSE: Disorders or differences of sex development (DSDs) are rare congenital conditions characterized by atypical sex development. Despite advances in genomic technologies, the molecular cause remains unknown in 50% of cases. METHODS: Homozygosity mapping and whole-exome sequencing revealed an ESR2 variant in an individual with syndromic 46,XY DSD. Additional cases with 46,XY DSD underwent whole-exome sequencing and targeted next-generation sequencing of ESR2. Functional characterization of the identified variants included luciferase assays and protein structure analysis. Gonadal ESR2 expression was assessed in human embryonic data sets and immunostaining of estrogen receptor-ß (ER-ß) was performed in an 8-week-old human male embryo. RESULTS: We identified a homozygous ESR2 variant, c.541_543del p.(Asn181del), located in the highly conserved DNA-binding domain of ER-ß, in an individual with syndromic 46,XY DSD. Two additional heterozygous missense variants, c.251G>T p.(Gly84Val) and c.1277T>G p.(Leu426Arg), located in the N-terminus and the ligand-binding domain of ER-ß, were found in unrelated, nonsyndromic 46,XY DSD cases. Significantly increased transcriptional activation and an impact on protein conformation were shown for the p.(Asn181del) and p.(Leu426Arg) variants. Testicular ESR2 expression was previously documented and ER-ß immunostaining was positive in the developing intestine and eyes. CONCLUSION: Our study supports a role for ESR2 as a novel candidate gene for 46,XY DSD.


Asunto(s)
Trastorno del Desarrollo Sexual 46,XY/genética , Receptor beta de Estrógeno/genética , Adolescente , Alelos , Sustitución de Aminoácidos/genética , Niño , Mapeo Cromosómico/métodos , Receptor beta de Estrógeno/metabolismo , Femenino , Frecuencia de los Genes/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Mutación/genética , Conformación Proteica , Relación Estructura-Actividad , Secuenciación del Exoma/métodos , Adulto Joven
17.
Sci Rep ; 7(1): 17211, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29222519

RESUMEN

The pro-inflammatory cytokine thymic stromal lymphopoietin (TSLP) plays a pivotal role in the pathophysiology of various allergy disorders that are mediated by type 2 helper T cell (Th2) responses, such as asthma and atopic dermatitis. TSLP forms a ternary complex with the TSLP receptor (TSLPR) and the interleukin-7-receptor subunit alpha (IL-7Rα), thereby activating a signaling cascade that culminates in the release of pro-inflammatory mediators. In this study, we conducted an in silico characterization of the TSLP:TSLPR complex to investigate the drugability of this complex. Two commercially available fragment libraries were screened computationally for possible inhibitors and a selection of fragments was subsequently tested in vitro. The screening setup consisted of two orthogonal assays measuring TSLP binding to TSLPR: a BLI-based assay and a biochemical assay based on a TSLP:alkaline phosphatase fusion protein. Four fragments pertaining to diverse chemical classes were identified to reduce TSLP:TSLPR complex formation to less than 75% in millimolar concentrations. We have used unbiased molecular dynamics simulations to develop a Markov state model that characterized the binding pathway of the most interesting compound. This work provides a proof-of-principle for use of fragments in the inhibition of TSLP:TSLPR complexation.


Asunto(s)
Citocinas/metabolismo , Receptores de Citocinas/metabolismo , Citocinas/química , Evaluación Preclínica de Medicamentos , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica/efectos de los fármacos , Conformación Proteica , Receptores de Citocinas/química , Células Th2/efectos de los fármacos , Células Th2/metabolismo , Interfaz Usuario-Computador , Linfopoyetina del Estroma Tímico
18.
Sci Rep ; 7(1): 15919, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162862

RESUMEN

STAT3 is a pleiotropic transcription factor involved in homeostatic and host defense processes in the human body. It is activated by numerous cytokines and growth factors and generates a series of cellular effects. Of the STAT-mediated signal transduction pathways, STAT3 transcriptional control is best understood. Jak kinase dependent activation of STAT3 relies on Y705 phosphorylation triggering a conformational switch that is stabilized by intermolecular interactions between SH2 domains and the pY705 motif. We here show that a second tyrosine phosphorylation within the SH2 domain at position Y640, induced by Tyk2, negatively controls STAT3 activity. The Y640F mutation leads to stabilization of activated STAT3 homodimers, accelerated nuclear translocation and superior transcriptional activity following IL-6 and LIF stimulation. Moreover, it unlocks type I IFN-dependent STAT3 signalling in cells that are normally refractory to STAT3 transcriptional activation.


Asunto(s)
Fosfotirosina/metabolismo , Factor de Transcripción STAT3/metabolismo , TYK2 Quinasa/metabolismo , Transcripción Genética , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Interferón-alfa/farmacología , Ratones , Proteínas Mutantes/metabolismo , Mutación/genética , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Multimerización de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Factor de Transcripción STAT3/genética , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , TYK2 Quinasa/química , Transcripción Genética/efectos de los fármacos
19.
PLoS One ; 12(5): e0178132, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28542518

RESUMEN

RNF41 (Ring Finger Protein 41) is an E3 ubiquitin ligase involved in the intracellular sorting and function of a diverse set of substrates. Next to BRUCE and Parkin, RNF41 can directly ubiquitinate ErbB3, IL-3, EPO and RARα receptors or downstream signaling molecules such as Myd88, TBK1 and USP8. In this way it can regulate receptor signaling and routing. To further elucidate the molecular mechanism behind the role of RNF41 in intracellular transport we performed an Array MAPPIT (Mammalian Protein-Protein Interaction Trap) screen using an extensive set of proteins derived from the human ORFeome collection. This paper describes the identification of VPS52, a subunit of the GARP (Golgi-Associated Retrograde Protein) and the EARP (Endosome-Associated Recycling Protein) complexes, as a novel interaction partner of RNF41. Through interaction via their coiled coil domains, RNF41 ubiquitinates and relocates VPS52 away from VPS53, a common subunit of the GARP and EARP complexes, towards RNF41 bodies.


Asunto(s)
Endosomas/metabolismo , Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/genética , Complejos Multiproteicos/genética , Ubiquitina-Proteína Ligasas/genética , Proteínas de Transporte Vesicular/genética
20.
Nat Commun ; 8: 14937, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28368013

RESUMEN

The pro-inflammatory cytokine thymic stromal lymphopoietin (TSLP) is pivotal to the pathophysiology of widespread allergic diseases mediated by type 2 helper T cell (Th2) responses, including asthma and atopic dermatitis. The emergence of human TSLP as a clinical target against asthma calls for maximally harnessing its therapeutic potential via structural and mechanistic considerations. Here we employ an integrative experimental approach focusing on productive and antagonized TSLP complexes and free cytokine. We reveal how cognate receptor TSLPR allosterically activates TSLP to potentiate the recruitment of the shared interleukin 7 receptor α-chain (IL-7Rα) by leveraging the flexibility, conformational heterogeneity and electrostatics of the cytokine. We further show that the monoclonal antibody Tezepelumab partly exploits these principles to neutralize TSLP activity. Finally, we introduce a fusion protein comprising a tandem of the TSLPR and IL-7Rα extracellular domains, which harnesses the mechanistic intricacies of the TSLP-driven receptor complex to manifest high antagonistic potency.


Asunto(s)
Asma/inmunología , Citocinas/antagonistas & inhibidores , Citocinas/química , Hipersensibilidad/inmunología , Complejos Multiproteicos/metabolismo , Receptores de Citocinas/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Asma/patología , Quimiocinas/biosíntesis , Cristalografía por Rayos X , Células Dendríticas , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Hipersensibilidad/patología , Modelos Moleculares , Estructura Secundaria de Proteína , Receptores de Citocinas/química , Receptores de Interleucina-7/química , Receptores de Interleucina-7/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Linfopoyetina del Estroma Tímico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...