Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Antimicrob Agents Chemother ; : e0002924, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38990015

RESUMO

Mycobacterium abscessus infections are emerging in cystic fibrosis patients, and treatment success rate in these patients is only 33% due to extreme antibiotic resistance. Thus, new treatment options are essential. An interesting target could be Lsr2, a nucleoid-associated protein involved in mycobacterial virulence. Zafirlukast is a Food and Drug Administration (FDA)-approved drug against asthma that was shown to bind Lsr2. In this study, zafirlukast treatment is shown to reduce M. abscessus growth, with a minimal inhibitory concentration of 16 µM and a bactericidal concentration of 64 µM in replicating bacteria only. As an initial response, DNA condensation, a known stress response of mycobacteria, occurs after 1 h of treatment with zafirlukast. During continued zafirlukast treatment, the morphology of the bacteria alters and the structural integrity of the bacteria is lost. After 4 days of treatment, reduced viability is measured in different culture media, and growth of M. abscessus is reduced in a dose-dependent manner. Using transmission electron microscopy, we demonstrated that the hydrophobic multilayered cell wall and periplasm are disorganized and ribosomes are reduced in size and relocalized. In summary, our data demonstrate that zafirlukast alters the morphology of M. abscessus and is bactericidal at 64 µM. The bactericidal concentration of zafirlukast is relatively high, and it is only effective on replicating bacteria but as zafirlukast is an FDA-approved drug, and currently used as an anti-asthma treatment, it could be an interesting drug to further study in in vivo experiments to determine whether it could be used as an antibiotic for M. abscessus infections.

2.
Mol Cell Proteomics ; 18(9): 1705-1720, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31138642

RESUMO

Huntington's disease is caused by a polyglutamine repeat expansion in the huntingtin protein which affects the function and folding of the protein, and results in intracellular protein aggregates. Here, we examined whether this mutation leads to altered ubiquitination of huntingtin and other proteins in both soluble and insoluble fractions of brain lysates of the Q175 knock-in Huntington's disease mouse model and the Q20 wild-type mouse model. Ubiquitination sites are detected by identification of Gly-Gly (diGly) remnant motifs that remain on modified lysine residues after digestion. We identified K6, K9, K132, K804, and K837 as endogenous ubiquitination sites of soluble huntingtin, with wild-type huntingtin being mainly ubiquitinated at K132, K804, and K837. Mutant huntingtin protein levels were strongly reduced in the soluble fraction whereas K6 and K9 were mainly ubiquitinated. In the insoluble fraction increased levels of huntingtin K6 and K9 diGly sites were observed for mutant huntingtin as compared with wild type. Besides huntingtin, proteins with various roles, including membrane organization, transport, mRNA processing, gene transcription, translation, catabolic processes and oxidative phosphorylation, were differently expressed or ubiquitinated in wild-type and mutant huntingtin brain tissues. Correlating protein and diGly site fold changes in the soluble fraction revealed that diGly site abundances of most of the proteins were not related to protein fold changes, indicating that these proteins were differentially ubiquitinated in the Q175 mice. In contrast, both the fold change of the protein level and diGly site level were increased for several proteins in the insoluble fraction, including ubiquitin, ubiquilin-2, sequestosome-1/p62 and myo5a. Our data sheds light on putative novel proteins involved in different cellular processes as well as their ubiquitination status in Huntington's disease, which forms the basis for further mechanistic studies to understand the role of differential ubiquitination of huntingtin and ubiquitin-regulated processes in Huntington's disease.


Assuntos
Encéfalo/metabolismo , Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Proteoma/metabolismo , Ubiquitina/metabolismo , Animais , Membrana Celular/metabolismo , Proteína Huntingtina/genética , Lisina/metabolismo , Camundongos Mutantes , Proteoma/análise , Solubilidade , Ubiquitinação , Fluxo de Trabalho
3.
Trends Immunol ; 37(11): 724-737, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27614798

RESUMO

Since the discovery of MHC molecules, it has taken 40 years to arrive at a coherent picture of how MHC class I and MHC class II molecules really work. This is a story of the proteases and MHC-like chaperones that support the MHC class I and II molecules in presenting peptides to the immune system. We now understand that the MHC system shapes both the repertoire of presented peptides and the subsequent T cell response, with important implications ranging from transplant rejection to tumor immunotherapies. Here we present an illustrated review of the ins and outs of MHC class I and MHC class II antigen presentation.


Assuntos
Apresentação de Antígeno , Rejeição de Enxerto/terapia , Antígenos de Histocompatibilidade Classe II/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoterapia/métodos , Neoplasias/terapia , Transplante de Órgãos , Linfócitos T/imunologia , Animais , Antígenos/imunologia , Rejeição de Enxerto/imunologia , Humanos , Imunoterapia/tendências , Ativação Linfocitária , Chaperonas Moleculares/metabolismo , Neoplasias/imunologia , Fragmentos de Peptídeos/imunologia , Peptídeo Hidrolases/metabolismo
4.
Hum Mol Genet ; 25(13): 2728-2737, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27260403

RESUMO

Spinocerebellar ataxia type 23 (SCA23) is caused by missense mutations in prodynorphin, encoding the precursor protein for the opioid neuropeptides α-neoendorphin, Dynorphin (Dyn) A and Dyn B, leading to neurotoxic elevated mutant Dyn A levels. Dyn A acts on opioid receptors to reduce pain in the spinal cord, but its cerebellar function remains largely unknown. Increased concentration of or prolonged exposure to Dyn A is neurotoxic and these deleterious effects are very likely caused by an N-methyl-d-aspartate-mediated non-opioid mechanism as Dyn A peptides were shown to bind NMDA receptors and potentiate their glutamate-evoked currents. In the present study, we investigated the cellular mechanisms underlying SCA23-mutant Dyn A neurotoxicity. We show that SCA23 mutations in the Dyn A-coding region disrupted peptide secondary structure leading to a loss of the N-terminal α-helix associated with decreased κ-opioid receptor affinity. Additionally, the altered secondary structure led to increased peptide stability of R6W and R9C Dyn A, as these peptides showed marked degradation resistance, which coincided with decreased peptide solubility. Notably, L5S Dyn A displayed increased degradation and no aggregation. R6W and wt Dyn A peptides were most toxic to primary cerebellar neurons. For R6W Dyn A, this is likely because of a switch from opioid to NMDA- receptor signalling, while for wt Dyn A, this switch was not observed. We propose that the pathology of SCA23 results from converging mechanisms of loss of opioid-mediated neuroprotection and NMDA-mediated excitotoxicity.


Assuntos
Dinorfinas/metabolismo , Degenerações Espinocerebelares/metabolismo , Sequência de Aminoácidos , Animais , Técnicas de Cultura de Células , Simulação por Computador , Dinorfinas/fisiologia , Endorfinas/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , N-Metilaspartato/metabolismo , Neurônios/metabolismo , Neurotoxinas , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Estrutura Secundária de Proteína , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Medula Espinal/metabolismo , Degenerações Espinocerebelares/genética
5.
Biol Chem ; 399(12): 1409-1419, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30138105

RESUMO

Histatins are histidine-rich peptides present in the saliva of humans and higher primates and have been implicated in the protection of the oral cavity. Histatin 1 is one of the most abundant histatins and recent reports show that it has a stimulating effect on cellular adherence, thereby suggesting a role in maintaining the quality of the epithelial barrier and stimulating mesenchymal-to-epithelial transition. Here we summarize these findings and discuss them in the context of previous reports. The recent findings also provide new insights in the physiological functions of histatin 1, which are discussed here. Furthermore, we put forward a possible role of histatin 1 in various pathologies and its potential function in clinical applications.


Assuntos
Transição Epitelial-Mesenquimal , Histatinas/metabolismo , Sequência de Aminoácidos , Adesão Celular , Histatinas/química , Histatinas/genética , Humanos
6.
Ophthalmic Res ; 59(2): 98-109, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29073609

RESUMO

INTRODUCTION: Curcumin has multiple biological effects including the modulation of protein homeostasis by the ubiquitin-proteasome system. The purpose of this study was to assess the in vitro cytotoxic and oxidative effects of nano-curcumin and standard curcumin and characterize their effects on proteasome regulation in retinal pigment epithelial (RPE) cells. METHODS: Viability, cell cycle progression, and reactive oxygen species (ROS) production were determined after treatment with nano-curcumin or curcumin. Subsequently, the effects of nano-curcumin and curcumin on proteasome activity and the gene and protein expression of proteasome subunits PA28α, α7, ß5, and ß5i were assessed. RESULTS: Nano-curcumin (5-100 µM) did not show significant cytotoxicity or anti-oxidative effects against H2O2-induced oxidative stress, whereas curcumin (≥10 µM) was cytotoxic and a potent inducer of ROS production. Both nano-curcumin and curcumin induced changes in proteasome-mediated proteolytic activity characterized by increased activity of the proteasome subunits ß2 and ß5i/ß1 and reduced activity of ß5/ß1i. Likewise, nano-curcumin and curcumin affected mRNA and protein levels of household and immunoproteasome subunits. CONCLUSIONS: Nano-curcumin is less toxic to RPE cells and less prone to induce ROS production than curcumin. Both nano-curcumin and curcumin increase proteasome-mediated proteolytic activity. These results suggest that nano-curcumin may be regarded as a proteasome-modulating agent of limited cytotoxicity for RPE cells.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Curcumina/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Pigmentos da Retina/metabolismo
7.
Glia ; 65(1): 50-61, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27615381

RESUMO

Huntington's disease (HD) is an autosomal dominant inherited neurodegenerative disorder that is caused by a CAG expansion in the Huntingtin (HTT) gene, leading to HTT inclusion formation in the brain. The mutant huntingtin protein (mHTT) is ubiquitously expressed and therefore nuclear inclusions could be present in all brain cells. The effects of nuclear inclusion formation have been mainly studied in neurons, while the effect on glia has been comparatively disregarded. Astrocytes, microglia, and oligodendrocytes are glial cells that are essential for normal brain function and are implicated in several neurological diseases. Here we examined the number of nuclear mHTT inclusions in both neurons and various types of glia in the two brain areas that are the most affected in HD, frontal cortex, and striatum. We compared nuclear mHTT inclusion body formation in three HD mouse models that express either full-length HTT or an N-terminal exon1 fragment of mHTT, and we observed nuclear inclusions in neurons, astrocytes, oligodendrocytes, and microglia. When studying the frequency of cells with nuclear inclusions in mice, we found that half of the population of neurons contained nuclear inclusions at the disease end stage, whereas the proportion of GFAP-positive astrocytes and oligodendrocytes having a nuclear inclusion was much lower, while microglia hardly showed any nuclear inclusions. Nuclear inclusions were also present in neurons and all studied glial cell types in human patient material. This is the first report to compare nuclear mHTT inclusions in glia and neurons in different HD mouse models and HD patient brains. GLIA 2016;65:50-61.


Assuntos
Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Animais , Astrócitos/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Doença de Huntington/metabolismo , Masculino , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo
8.
Biochim Biophys Acta Mol Basis Dis ; 1863(3): 793-800, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28040507

RESUMO

Most neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's disease are hallmarked by aggregate formation of disease-related proteins. In various of these diseases transfer of aggregation-prone proteins between neurons and between neurons and glial cells has been shown, thereby initiating aggregation in neighboring cells and so propagating the disease phenotype. Whereas this prion-like transfer is well studied in Alzheimer's and Parkinson's disease, only a few studies have addressed this potential mechanism in Huntington's disease. Here, we present an overview of in vitro and in vivo methodologies to study release, intercellular transfer and uptake of aggregation-prone protein fragments in Huntington's disease models.


Assuntos
Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Príons/metabolismo , Agregação Patológica de Proteínas/metabolismo , Animais , Humanos , Proteína Huntingtina/análise , Proteína Huntingtina/genética , Doença de Huntington/genética , Doença de Huntington/patologia , Mutação , Neuroglia/patologia , Neurônios/patologia , Príons/análise , Príons/genética , Agregados Proteicos , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia , Transporte Proteico
9.
Cell Mol Life Sci ; 73(21): 4101-20, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27141937

RESUMO

Glial fibrillary acidic protein (GFAP) is the characteristic intermediate filament (IF) protein in astrocytes. Expression of its main isoforms, GFAPα and GFAPδ, varies in astrocytes and astrocytoma implying a potential regulatory role in astrocyte physiology and pathology. An IF-network is a dynamic structure and has been functionally linked to cell motility, proliferation, and morphology. There is a constant exchange of IF-proteins with the network. To study differences in the dynamic properties of GFAPα and GFAPδ, we performed fluorescence recovery after photobleaching experiments on astrocytoma cells with fluorescently tagged GFAPs. Here, we show for the first time that the exchange of GFP-GFAPδ was significantly slower than the exchange of GFP-GFAPα with the IF-network. Furthermore, a collapsed IF-network, induced by GFAPδ expression, led to a further decrease in fluorescence recovery of both GFP-GFAPα and GFP-GFAPδ. This altered IF-network also changed cell morphology and the focal adhesion size, but did not alter cell migration or proliferation. Our study provides further insight into the modulation of the dynamic properties and functional consequences of the IF-network composition.


Assuntos
Astrócitos/citologia , Forma Celular , Adesões Focais/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Filamentos Intermediários/metabolismo , Actinas/metabolismo , Adulto , Idoso , Astrócitos/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Feminino , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imageamento Tridimensional , Microtúbulos/metabolismo , Nestina/metabolismo , Isoformas de Proteínas/metabolismo , Vimentina/metabolismo
10.
Mol Cell Proteomics ; 14(8): 2177-93, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26041847

RESUMO

Tripeptidyl peptidase II (TPP2) is a serine peptidase involved in various biological processes, including antigen processing, cell growth, DNA repair, and neuropeptide mediated signaling. The underlying mechanisms of how a peptidase can influence this multitude of processes still remain unknown. We identified rapid proteomic changes in neuroblastoma cells following selective TPP2 inhibition using the known reversible inhibitor butabindide, as well as a new, more potent, and irreversible peptide phosphonate inhibitor. Our data show that TPP2 inhibition indirectly but rapidly decreases the levels of active, di-phosphorylated extracellular signal-regulated kinase 1 (ERK1) and ERK2 in the nucleus, thereby down-regulating signal transduction downstream of growth factors and mitogenic stimuli. We conclude that TPP2 mediates many important cellular functions by controlling ERK1 and ERK2 phosphorylation. For instance, we show that TPP2 inhibition of neurons in the hippocampus leads to an excessive strengthening of synapses, indicating that TPP2 activity is crucial for normal brain function.


Assuntos
Aminopeptidases/metabolismo , Núcleo Celular/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Serina Endopeptidases/metabolismo , Aminopeptidases/antagonistas & inibidores , Animais , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Dipeptidil Peptidases e Tripeptidil Peptidases/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Técnicas de Silenciamento de Genes , Ontologia Genética , Humanos , Concentração Inibidora 50 , Marcação por Isótopo , Camundongos , Modelos Biológicos , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 2/metabolismo , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXC/genética , Fatores de Transcrição SOXC/metabolismo
11.
J Biol Chem ; 288(38): 27068-27084, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23908352

RESUMO

Huntington disease is a neurodegenerative disorder caused by an expanded polyglutamine (polyQ) repeat within the protein huntingtin (Htt). N-terminal fragments of the mutant Htt (mHtt) proteins containing the polyQ repeat are aggregation-prone and form intracellular inclusion bodies. Improving the clearance of mHtt fragments by intracellular degradation pathways is relevant to obviate toxic mHtt species and subsequent neurodegeneration. Because the proteasomal degradation pathway has been the subject of controversy regarding the processing of expanded polyQ repeats, we examined whether the proteasome can efficiently degrade Htt-exon1 with an expanded polyQ stretch both in neuronal cells and in vitro. Upon targeting mHtt-exon1 to the proteasome, rapid and complete clearance of mHtt-exon1 was observed. Proteasomal degradation of mHtt-exon1 was devoid of polyQ peptides as partial cleavage products by incomplete proteolysis, indicating that mammalian proteasomes are capable of efficiently degrading expanded polyQ sequences without an inhibitory effect on the proteasomal activity.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Animais , Linhagem Celular , Humanos , Proteína Huntingtina , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Peptídeos/genética , Complexo de Endopeptidases do Proteassoma/genética , Sequências Repetitivas de Aminoácidos
12.
J Biol Chem ; 288(24): 17225-37, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23612975

RESUMO

Fragments of proteins containing an expanded polyglutamine (polyQ) tract are thought to initiate aggregation and toxicity in at least nine neurodegenerative diseases, including Huntington's disease. Because proteasomes appear unable to digest the polyQ tract, which can initiate intracellular protein aggregation, preventing polyQ peptide aggregation by chaperones should greatly improve polyQ clearance and prevent aggregate formation. Here we expressed polyQ peptides in cells and show that their intracellular aggregation is prevented by DNAJB6 and DNAJB8, members of the DNAJ (Hsp40) chaperone family. In contrast, HSPA/Hsp70 and DNAJB1, also members of the DNAJ chaperone family, did not prevent peptide-initiated aggregation. Intriguingly, DNAJB6 and DNAJB8 also affected the soluble levels of polyQ peptides, indicating that DNAJB6 and DNAJB8 inhibit polyQ peptide aggregation directly. Together with recent data showing that purified DNAJB6 can suppress fibrillation of polyQ peptides far more efficiently than polyQ expanded protein fragments in vitro, we conclude that the mechanism of DNAJB6 and DNAJB8 is suppression of polyQ protein aggregation by directly binding the polyQ tract.


Assuntos
Proteínas de Choque Térmico HSP40/fisiologia , Chaperonas Moleculares/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fragmentos de Peptídeos/metabolismo , Peptídeos/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HEK293 , Células HeLa , Humanos , Ligação Proteica , Multimerização Proteica , Solubilidade
13.
J Neurochem ; 128(5): 741-51, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24134140

RESUMO

The protein kinase C γ (PKCγ) undergoes multistep activation and participates in various cellular processes in Purkinje cells. Perturbations in its phosphorylation state, conformation or localization can disrupt kinase signalling, such as in spinocerebellar ataxia type 14 (SCA14) that is caused by missense mutations in PRKCG encoding for PKCγ. We previously showed that SCA14 mutations enhance PKCγ membrane translocation upon stimulation owing to an altered protein conformation. As the faster translocation did not result in an increased function, we examined how SCA14 mutations induce this altered conformation of PKCγ and what the consequences of this conformational change are on PKCγ life cycle. Here, we show that SCA14-related PKCγ-V138E exhibits an exposed C-terminus as shown by fluorescence resonance energy transfer-fluorescence lifetime imaging microscopy in living cells, indicative of its partial unfolding. This conformational change was associated with faster phorbol 12-myristate 13-acetate-induced translocation and accumulation of fully phosphorylated PKCγ in the insoluble fraction, which could be rescued by coexpressing PDK1 kinase that normally triggers PKCγ autophosphorylation. We propose that the SCA14 mutation V138E causes unfolding of the C1B domain and exposure of the C-terminus of the PKCγ-V138E molecule, resulting in a decrease of functional kinase in the soluble fraction. Here, we show that the mutation V138E of the protein kinase C γ (PKCγ) C1B domain (PKCγ-V138E), which is implicated in spinocerebellar ataxia type 14, exhibits a partially unfolded C-terminus. This leads to unusually fast phorbol 12-myristate 13-acetate-induced membrane translocation and accumulation of phosphorylated PKCγ-V138E in the insoluble fraction, causing loss of the functional kinase. In contrast to general chaperones, coexpression of PKCγ's 'natural chaperone', PDK1 kinase, could rescue the PKCγ-V138E phenotype.


Assuntos
Proteína Quinase C/genética , Animais , Western Blotting , Células COS , Carcinógenos/farmacologia , Chlorocebus aethiops , DNA/genética , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Cinética , Mutação de Sentido Incorreto/genética , Mutação de Sentido Incorreto/fisiologia , Fosforilação , Polietilenoglicóis/química , Dobramento de Proteína , Proteína Quinase C/química , Proteínas Serina-Treonina Quinases/biossíntese , Piruvato Desidrogenase Quinase de Transferência de Acetil , Solubilidade , Solventes , Ataxias Espinocerebelares/genética , Acetato de Tetradecanoilforbol/farmacologia
14.
J Huntingtons Dis ; 13(2): 201-214, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38640164

RESUMO

Background: Huntington's disease is an inheritable autosomal dominant disorder caused by an expanded CAG trinucleotide repeat within the Huntingtin gene, leading to a polyglutamine (polyQ) expansion in the mutant protein. Objective: A potential therapeutic approach for delaying or preventing the onset of the disease involves enhancing the degradation of the aggregation-prone polyQ-expanded N-terminal mutant huntingtin (mHTT) exon1 fragment. A few proteases and peptidases have been identified that are able to cleave polyQ fragments with low efficiency. This study aims to identify a potent polyQ-degrading endopeptidase. Methods: Here we used quenched polyQ peptides to identify a polyQ-degrading endopeptidase. Next we investigated its role on HTT turnover, using purified polyQ-expanded HTT fragments and striatal cells expressing mHTT exon1 peptides. Results: We identified insulin-degrading enzyme (IDE) as a novel endopeptidase for degrading polyQ peptides. IDE was, however, ineffective in reducing purified polyQ-expanded HTT fragments. Similarly, in striatal cells expressing mHTT exon1 peptides, IDE did not enhance mHTT turnover. Conclusions: This study shows that despite IDE's efficiency in degrading polyQ peptides, it does not contribute to the direct degradation of polyQ-expanded mHTT fragments.


Assuntos
Proteína Huntingtina , Insulisina , Peptídeos , Insulisina/metabolismo , Insulisina/genética , Proteína Huntingtina/metabolismo , Proteína Huntingtina/genética , Peptídeos/metabolismo , Humanos , Animais , Doença de Huntington/metabolismo , Doença de Huntington/genética , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Corpo Estriado/metabolismo
15.
Traffic ; 12(12): 1897-910, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21883763

RESUMO

Most cytoplasmic and nuclear proteins are degraded via the ubiquitin-proteasome system into peptides, which are subsequently hydrolyzed by downstream aminopeptidases. Inefficient degradation can lead to accumulation of protein fragments, and subsequent aggregation and toxicity. Whereas the role of the proteasome and the effect of its impairment on aggregation have been intensively studied, little is known about how cells deal with peptides that show resistance to degradation by aminopeptidases. Here, we introduced peptidase-resistant peptides into living cells and show that these peptides rapidly and irreversibly accumulate into puncta in the perinuclear region of the cell. Accumulation appears to be independent of peptide sequence but is less efficient for longer peptides. The puncta colocalize with autophagosomal and lysosomal markers, suggesting that these peptides end up within lysosomes via macroautophagy. Surprisingly, the peptides still accumulate within lysosomes when macroautophagy is impaired, suggesting a trafficking route independent of macroautophagy. Upon lysosomal uptake, peptides are degraded, suggesting that cells can clear peptidase-resistant proteasomal products by an alternative pathway, which targets them to lysosomes.


Assuntos
Aminopeptidases/metabolismo , Lisossomos/metabolismo , Peptídeos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Citoplasma/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Células HeLa , Humanos , Melanoma/metabolismo , Fagossomos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Isoformas de Proteínas , Proteólise
16.
Blood ; 118(19): 5178-88, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21948297

RESUMO

Sensitivity of chronic lymphocytic leukemia (CLL) cells to anti-CD20 mAbs is low and, therefore, the efficacy of monotherapy with current anti-CD20 mAbs is limited. At present, it is not known whether sensitivity of CLL cells to CD20 mAbs is modulated by microenvironmental stimuli. We have shown previously that in vitro CD40 stimulation of peripheral blood-derived CLL cells results in resistance to cytotoxic drugs. In the present study, we show that, in contrast, CD40 stimulation sensitizes CLL cells to the recently described novel type II anti-CD20 mAb GA101. Cell death occurred without cross-linking of GA101 and involved a lysosome-dependent mechanism. Combining GA101 with various cytotoxic drugs resulted in additive cell death, not only in CD40-stimulated CLL cells, but also in p53-dysfunctional CLL cells. Our findings indicate that GA101 has efficacy against chemoresistant CLL, and provide a rationale for combining cytotoxic drugs with anti-CD20 mAbs.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos CD40/metabolismo , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/terapia , Actinas/metabolismo , Adulto , Idoso , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Murinos/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Morte Celular/imunologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Técnicas In Vitro , Leucemia Linfocítica Crônica de Células B/metabolismo , Leucemia Linfocítica Crônica de Células B/patologia , Lisossomos/imunologia , Masculino , Pessoa de Meia-Idade , Rituximab
17.
Front Mol Biosci ; 10: 1107323, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36926679

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the N-terminus of the HTT gene. The CAG repeat expansion translates into a polyglutamine expansion in the mutant HTT (mHTT) protein, resulting in intracellular aggregation and neurotoxicity. Lowering the mHTT protein by reducing synthesis or improving degradation would delay or prevent the onset of HD, and the ubiquitin-proteasome system (UPS) could be an important pathway to clear the mHTT proteins prior to aggregation. The UPS is not impaired in HD, and proteasomes can degrade mHTT entirely when HTT is targeted for degradation. However, the mHTT protein is differently ubiquitinated when compared to wild-type HTT (wtHTT), suggesting that the polyQ expansion affects interaction with (de) ubiquitinating enzymes and subsequent targeting for degradation. The soluble mHTT protein is associated with several ubiquitin-modifying enzymes, and various ubiquitin-modifying enzymes have been identified that are linked to Huntington's disease, either by improving mHTT turnover or affecting overall homeostasis. Here we describe their potential mechanism of action toward improved mHTT targeting towards the proteostasis machinery.

18.
Microbiol Spectr ; 11(6): e0302923, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37975661

RESUMO

IMPORTANCE: Even though the coronavirus disease 2019 (COVID-19) pandemic is slowly developing into a conventional infectious disease, the long-term effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infection are still not well understood. One of the problems is that many COVID-19 cases develop acute kidney injuries. Still, it is heavily debated whether SARS-CoV-2 virus enters and actively replicates in kidney tissue and if SARS-CoV-2 virus particles can be detected in kidney during or post-infection. Here, we demonstrated that nucleocapsid N protein was detected in kidney tubular epithelium of patients that already recovered form COVID-19. The presence of the abundantly produced N protein without signs of viral replication could have implications for the recurrence of kidney disease and have a continuing effect on the immune system.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Proteínas do Nucleocapsídeo , Replicação Viral , Epitélio
19.
J Biol Chem ; 286(34): 30190-9, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21734303

RESUMO

We recently identified the liver X receptor-regulated E3 ubiquitin ligase inducible degrader of the LDL receptor (IDOL) as a modulator of lipoprotein metabolism. Acting as an E3 ubiquitin ligase, IDOL triggers ubiquitination and subsequent degradation of the low density lipoprotein receptor (LDLR). We demonstrate here that this outcome requires the conserved FERM and RING domains present in IDOL. The RING domain promotes ubiquitination in vitro and Lys-63-specific ubiquitination of the LDLR in vivo in response to IDOL or liver X receptor activation. We further identify RING residues that differentially influence ubiquitination of the LDLR or stability of IDOL. The FERM domain interacts with the LDLR and in living cells co-localizes with the receptor at the plasma membrane. Homology modeling revealed a phosphotyrosine-binding element embedded in the FERM domain. Mutating residues within this region or residues in the LDLR preceding the NPVY endocytosis motif abrogate LDLR degradation by IDOL. Collectively, our results indicate that both the FERM and RING domains are required for promoting lysosomal degradation of the LDLR by IDOL. Our findings may facilitate development of structure-based IDOL inhibitors aimed at increasing LDLR abundance in therapeutic strategies to treat cardiovascular disease.


Assuntos
Lisossomos/metabolismo , Receptores de LDL/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/fisiologia , Motivos de Aminoácidos , Células HEK293 , Células Hep G2 , Humanos , Receptores X do Fígado , Lisossomos/genética , Mutação , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Domínios RING Finger , Receptores de LDL/genética , Ubiquitina-Proteína Ligases/genética
20.
J Exp Med ; 203(5): 1259-71, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16636135

RESUMO

Radiotherapy is one of the most successful cancer therapies. Here the effect of irradiation on antigen presentation by MHC class I molecules was studied. Cell surface expression of MHC class I molecules was increased for many days in a radiation dose-dependent manner as a consequence of three responses. Initially, enhanced degradation of existing proteins occurred which resulted in an increased intracellular peptide pool. Subsequently, enhanced translation due to activation of the mammalian target of rapamycin pathway resulted in increased peptide production, antigen presentation, as well as cytotoxic T lymphocyte recognition of irradiated cells. In addition, novel proteins were made in response to gamma-irradiation, resulting in new peptides presented by MHC class I molecules, which were recognized by cytotoxic T cells. We show that immunotherapy is successful in eradicating a murine colon adenocarcinoma only when preceded by radiotherapy of the tumor tissue. Our findings indicate that directed radiotherapy can improve the efficacy of tumor immunotherapy.


Assuntos
Adenocarcinoma/imunologia , Apresentação de Antígeno/efeitos da radiação , Neoplasias do Colo/imunologia , Raios gama , Antígeno HLA-A2/imunologia , Imunoterapia , Adenocarcinoma/genética , Adenocarcinoma/terapia , Animais , Apresentação de Antígeno/imunologia , Neoplasias do Colo/genética , Neoplasias do Colo/terapia , Relação Dose-Resposta à Radiação , Regulação Neoplásica da Expressão Gênica/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Antígeno HLA-A2/genética , Humanos , Camundongos , Camundongos Transgênicos , Peptídeos/imunologia , Biossíntese de Proteínas/imunologia , Biossíntese de Proteínas/efeitos da radiação , Proteínas Quinases/imunologia , Radioterapia , Linfócitos T Citotóxicos/imunologia , Serina-Treonina Quinases TOR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA