Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Life Sci Alliance ; 6(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37553253

RESUMO

N-terminal phosphorylation at residues T3 and S13 is believed to have important beneficial implications for the biological and pathological properties of mutant huntingtin, where inhibitor of nuclear factor kappa B kinase subunit beta (IKBKB) was identified as a candidate regulator of huntingtin N-terminal phosphorylation. The paucity of mechanistic information on IKK pathways, together with the lack of sensitive methods to quantify endogenous huntingtin phosphorylation, prevented detailed study of the role of IKBKB in Huntington's disease. Using novel ultrasensitive assays, we demonstrate that IKBKB can regulate endogenous S13 huntingtin phosphorylation in a manner, dependent on its kinase activity and known regulators. We found that the ability of IKBKB to phosphorylate endogenous huntingtin S13 is mediated through a non-canonical interferon regulatory factor3-mediated IKK pathway, distinct from the established involvement of IKBKB in mutant huntingtin's pathological mechanisms mediated via the canonical pathway. Furthermore, increased huntingtin S13 phosphorylation by IKBKB resulted in decreased aggregation of mutant huntingtin in cells, again dependent on its kinase activity. These findings point to a non-canonical IKK pathway linking S13 huntingtin phosphorylation to the pathological properties of mutant huntingtin aggregation, thought to be significant to Huntington's disease.


Assuntos
Doença de Huntington , Quinase I-kappa B , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Serina/metabolismo , Fosforilação
2.
EMBO J ; 39(17): e104671, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32757223

RESUMO

Phosphorylation of the N-terminal domain of the huntingtin (HTT) protein has emerged as an important regulator of its localization, structure, aggregation, clearance and toxicity. However, validation of the effect of bona fide phosphorylation in vivo and assessing the therapeutic potential of targeting phosphorylation for the treatment of Huntington's disease (HD) require the identification of the enzymes that regulate HTT phosphorylation. Herein, we report the discovery and validation of a kinase, TANK-binding kinase 1 (TBK1), that efficiently phosphorylates full-length and N-terminal HTT fragments in vitro (at S13/S16), in cells (at S13) and in vivo. TBK1 expression in HD models (cells, primary neurons, and Caenorhabditis elegans) increases mutant HTT exon 1 phosphorylation and reduces its aggregation and cytotoxicity. We demonstrate that the TBK1-mediated neuroprotective effects are due to phosphorylation-dependent inhibition of mutant HTT exon 1 aggregation and an increase in autophagic clearance of mutant HTT. These findings suggest that upregulation and/or activation of TBK1 represents a viable strategy for the treatment of HD by simultaneously lowering mutant HTT levels and blocking its aggregation.


Assuntos
Caenorhabditis elegans/metabolismo , Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Mutação , Agregados Proteicos , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Modelos Animais de Doenças , Células HEK293 , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Ratos
3.
Biochem Biophys Res Commun ; 521(3): 549-554, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31677786

RESUMO

Huntington's disease (HD) is a progressive neurodegenerative disorder caused by an expansion of a CAG triplet repeat (encoding for a polyglutamine tract) within the first exon of the huntingtin gene. Expression of the mutant huntingtin (mHTT) protein can result in the production of N-terminal fragments with a robust propensity to form oligomers and aggregates, which may be causally associated with HD pathology. Several lines of evidence indicate that N17 phosphorylation or pseudophosphorylation at any of the residues T3, S13 or S16, alone or in combination, modulates mHTT aggregation, subcellular localization and toxicity. Consequently, increasing N17 phosphorylation has been proposed as a potential therapeutic approach. However, developing genetic/pharmacological tools to quantify these phosphorylation events is necessary in order to subsequently develop tool modulators, which is difficult given the transient and incompletely penetrant nature of such post-translational modifications. Here we describe the first ultrasensitive sandwich immunoassay that quantifies HTT phosphorylated at residue S13 and demonstrate its utility for specific analyte detection in preclinical models of HD.


Assuntos
Proteína Huntingtina/análise , Animais , Células Cultivadas , Técnicas de Introdução de Genes , Células HEK293 , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Camundongos , Mutação , Neurônios/química , Neurônios/metabolismo , Fosforilação , Agregados Proteicos , Processamento de Proteína Pós-Traducional
4.
Front Neurosci ; 13: 889, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31507364

RESUMO

Accumulation and aggregation of misfolded alpha-synuclein is believed to be a cause of Parkinson's disease (PD). Phosphorylation of alpha-synuclein at S129 is known to be associated with the pathological misfolding process, but efforts to investigate the relevance of this post-translational modification for pathology have been frustrated by difficulties in detecting and quantifying it in relevant samples. We report novel, ultrasensitive immunoassays based on single-molecule counting technology, useful for detecting alpha-synuclein and its S129 phosphorylated form in clinical samples in the low pg/ml range. Using human CSF and plasma samples, we find levels of alpha-synuclein comparable to those previously reported. However, while alpha-synuclein phosphorylated on S129 could easily be detected in human plasma, where its detection is extremely sensitive to protein phosphatases, its levels in CSF were undetectable, with a possible influence of a matrix effect. In plasma samples from a small test cohort comprising 5 PD individuals and five age-matched control individuals we find that pS129 alpha-synuclein levels are increased in PD plasma samples, in line with previous reports. We conclude that pS129 alpha-synuclein is not detectable in CSF and recommend the addition of phosphatase inhibitors to plasma samples at the time of collection. Moreover, the findings obtained on the small cohort of clinical plasma samples point to plasma pS129 alpha-synuclein levels as a candidate diagnostic biomarker in PD.

5.
J Clin Invest ; 129(6): 2390-2403, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-31063986

RESUMO

A disintegrine and metalloproteinase 10 (ADAM10) is implicated in synaptic function through its interaction with postsynaptic receptors and adhesion molecules. Here, we report that levels of active ADAM10 are increased in Huntington's disease (HD) mouse cortices and striata and in human postmortem caudate. We show that, in the presence of polyglutamine-expanded (polyQ-expanded) huntingtin (HTT), ADAM10 accumulates at the postsynaptic densities (PSDs) and causes excessive cleavage of the synaptic protein N-cadherin (N-CAD). This aberrant phenotype is also detected in neurons from HD patients where it can be reverted by selective silencing of mutant HTT. Consistently, ex vivo delivery of an ADAM10 synthetic inhibitor reduces N-CAD proteolysis and corrects electrophysiological alterations in striatal medium-sized spiny neurons (MSNs) of 2 HD mouse models. Moreover, we show that heterozygous conditional deletion of ADAM10 or delivery of a competitive TAT-Pro-ADAM10709-729 peptide in R6/2 mice prevents N-CAD proteolysis and ameliorates cognitive deficits in the mice. Reduction in synapse loss was also found in R6/2 mice conditionally deleted for ADAM10. Taken together, these results point to a detrimental role of hyperactive ADAM10 at the HD synapse and provide preclinical evidence of the therapeutic potential of ADAM10 inhibition in HD.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Disfunção Cognitiva/enzimologia , Doença de Huntington/enzimologia , Proteínas de Membrana/metabolismo , Densidade Pós-Sináptica/enzimologia , Proteína ADAM10/genética , Adulto , Idoso , Secretases da Proteína Precursora do Amiloide/genética , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Doença de Huntington/genética , Doença de Huntington/patologia , Masculino , Proteínas de Membrana/genética , Camundongos Transgênicos , Pessoa de Meia-Idade , Densidade Pós-Sináptica/genética , Densidade Pós-Sináptica/patologia
6.
Proc Natl Acad Sci U S A ; 114(50): E10809-E10818, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29162692

RESUMO

Posttranslational modifications can have profound effects on the biological and biophysical properties of proteins associated with misfolding and aggregation. However, their detection and quantification in clinical samples and an understanding of the mechanisms underlying the pathological properties of misfolding- and aggregation-prone proteins remain a challenge for diagnostics and therapeutics development. We have applied an ultrasensitive immunoassay platform to develop and validate a quantitative assay for detecting a posttranslational modification (phosphorylation at residue T3) of a protein associated with polyglutamine repeat expansion, namely Huntingtin, and characterized its presence in a variety of preclinical and clinical samples. We find that T3 phosphorylation is greatly reduced in samples from Huntington's disease models and in Huntington's disease patients, and we provide evidence that bona-fide T3 phosphorylation alters Huntingtin exon 1 protein conformation and aggregation properties. These findings have significant implications for both mechanisms of disease pathogenesis and the development of therapeutics and diagnostics for Huntington's disease.


Assuntos
Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Imunoensaio/métodos , Animais , Células Cultivadas , Éxons , Células HEK293 , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Camundongos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilação , Conformação Proteica , Processamento de Proteína Pós-Traducional , Sensibilidade e Especificidade
7.
Sci Rep ; 7(1): 5070, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28698602

RESUMO

Conformational changes in disease-associated or mutant proteins represent a key pathological aspect of Huntington's disease (HD) and other protein misfolding diseases. Using immunoassays and biophysical approaches, we and others have recently reported that polyglutamine expansion in purified or recombinantly expressed huntingtin (HTT) proteins affects their conformational properties in a manner dependent on both polyglutamine repeat length and temperature but independent of HTT protein fragment length. These findings are consistent with the HD mutation affecting structural aspects of the amino-terminal region of the protein, and support the concept that modulating mutant HTT conformation might provide novel therapeutic and diagnostic opportunities. We now report that the same conformational TR-FRET based immunoassay detects polyglutamine- and temperature-dependent changes on the endogenously expressed HTT protein in peripheral tissues and post-mortem HD brain tissue, as well as in tissues from HD animal models. We also find that these temperature- and polyglutamine-dependent conformational changes are sensitive to bona-fide phosphorylation on S13 and S16 within the N17 domain of HTT. These findings provide key clinical and preclinical relevance to the conformational immunoassay, and provide supportive evidence for its application in the development of therapeutics aimed at correcting the conformation of polyglutamine-expanded proteins as well as the pharmacodynamics readouts to monitor their efficacy in preclinical models and in HD patients.


Assuntos
Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Peptídeos/metabolismo , Expansão das Repetições de Trinucleotídeos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Drosophila/metabolismo , Éxons/genética , Fibroblastos/metabolismo , Células HEK293 , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilação , Fosfosserina/metabolismo , Conformação Proteica
8.
Biochem Biophys Res Commun ; 478(2): 949-55, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27520369

RESUMO

We have previously reported TR-FRET based immunoassays to detect a conformational change imparted on huntingtin protein by the polyglutamine expansion, which we confirmed using biophysical methodologies. Using these immunoassays, we now report that polyglutamine expansion influences the conformational properties of other polyglutamine disease proteins, exemplified by the androgen receptor (associated with spinal bulbar muscular atrophy) and TATA binding protein (associated with spinocerebellar ataxia 17). Using artificial constructs bearing short or long polyglutamine expansions or a multimerized, unrelated epitope (mimicking the increase in anti-polyglutamine antibody epitopes present in polyglutamine repeats of increasing length) we confirmed that the conformational TR-FRET based immunoassay detects an intrinsic conformational property of polyglutamine repeats. The TR-FRET based conformational immunoassay may represent a rapid, scalable tool to identify modulators of polyglutamine-mediated conformational change in different proteins associated with CAG triplet repeat disorders.


Assuntos
Doença/genética , Conformação Molecular , Peptídeos/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Extratos Celulares , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Imunoensaio , Transfecção
9.
PLoS One ; 10(7): e0132617, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26147970

RESUMO

Regulatory T cells (Tregs) are physiologically designed to prevent autoimmune disease and maintain self-tolerance. In tumour microenvironments, their presence is related to a poor prognosis, and they influence the therapeutic outcome due to their capacity to suppress the immune response by cell-cell contact and to release immunosuppressive cytokines. In this study, we demonstrate that Treg immunosuppressive activity can be modulated by the cross-linking between the CD45RA expressed by Tregs and the C-type lectin MGL. This specific interaction strongly decreases the immunosuppressive activity of Tregs, restoring the proliferative capacity of co-cultured T lymphocytes. This effect can be attributed to changes in CD45RA and TCR signalling through the inhibition of Lck and inactivation of Zap-70, an increase in the Foxp3 methylation status and, ultimately, the reduced production of suppressive cytokines. These results indicate a role of MGL as an immunomodulator within the tumour microenvironment interfering with Treg functions, suggesting its possible use in the design of anticancer vaccines.


Assuntos
Tolerância Imunológica , Lectinas Tipo C/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Técnicas de Cocultura , Fatores de Transcrição Forkhead/imunologia , Humanos , Antígenos Comuns de Leucócito/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Reguladores/citologia , Proteína-Tirosina Quinase ZAP-70/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA