Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochem J ; 478(24): 4203-4220, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34821356

RESUMO

SLC26A9, a member of the solute carrier protein family, transports chloride ions across various epithelia. SLC26A9 also associates with other ion channels and transporters linked to human health, and in some cases these heterotypic interactions are essential to support the biogenesis of both proteins. Therefore, understanding how this complex membrane protein is initially folded might provide new therapeutic strategies to overcome deficits in the function of SLC26A9 partners, one of which is associated with Cystic Fibrosis. To this end, we developed a novel yeast expression system for SLC26A9. This facile system has been used extensively with other ion channels and transporters to screen for factors that oversee protein folding checkpoints. As commonly observed for other channels and transporters, we first noted that a substantial fraction of SLC26A9 is targeted for endoplasmic reticulum associated degradation (ERAD), which destroys folding-compromised proteins in the early secretory pathway. We next discovered that ERAD selection requires the Hsp70 chaperone, which can play a vital role in ERAD substrate selection. We then created SLC26A9 mutants and found that the transmembrane-rich domain of SLC26A9 was quite stable, whereas the soluble cytosolic STAS domain was responsible for Hsp70-dependent ERAD. To support data obtained in the yeast model, we were able to recapitulate Hsp70-facilitated ERAD of the STAS domain in human tissue culture cells. These results indicate that a critical barrier to nascent membrane protein folding can reside within a specific soluble domain, one that is monitored by components associated with the ERAD machinery.


Assuntos
Antiporters/metabolismo , Degradação Associada com o Retículo Endoplasmático , Proteínas de Choque Térmico HSP70/metabolismo , Modelos Biológicos , Transportadores de Sulfato/metabolismo , Antiporters/genética , Células HEK293 , Proteínas de Choque Térmico HSP70/genética , Humanos , Domínios Proteicos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transportadores de Sulfato/genética
2.
Bioorg Med Chem Lett ; 48: 128243, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34246753

RESUMO

A growing number of diseases are linked to the misfolding of integral membrane proteins, and many of these proteins are targeted for ubiquitin-proteasome-dependent degradation. One such substrate is a mutant form of the Cystic Fibrosis Transmembrane Conductance Regulator (F508del-CFTR). Protein folding "correctors" that repair the F508del-CFTR folding defect have entered the clinic, but they are unlikely to protect the entire protein from degradation. To increase the pool of F508del-CFTR protein that is available for correction by existing treatments, we determined a structure-activity relationship to improve the efficacy and reduce the toxicity of an inhibitor of the E1 ubiquitin activating enzyme that facilitates F508del-CFTR maturation. A resulting lead compound lacked measurable toxicity and improved the ability of an FDA-approved corrector to augment F508del-CFTR folding, transport the protein to the plasma membrane, and maintain its activity. These data support a proof-of-concept that modest inhibition of substrate ubiquitination improves the activity of small molecule correctors to treat CF and potentially other protein conformational disorders.


Assuntos
Benzoatos/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Furanos/farmacologia , Pirazóis/farmacologia , Ubiquitina/antagonistas & inibidores , Benzoatos/química , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Relação Dose-Resposta a Droga , Furanos/química , Humanos , Estrutura Molecular , Dobramento de Proteína/efeitos dos fármacos , Pirazóis/química , Relação Estrutura-Atividade , Ubiquitina/metabolismo , Ubiquitinação/efeitos dos fármacos
3.
Bioorg Med Chem ; 27(1): 79-91, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30528127

RESUMO

Over-expression of the Hsp70 molecular chaperone prevents protein aggregation and ameliorates neurodegenerative disease phenotypes in model systems. We identified an Hsp70 activator, MAL1-271, that reduces α-synuclein aggregation in a Parkinson's Disease model. We now report that MAL1-271 directly increases the ATPase activity of a eukaryotic Hsp70. Next, twelve MAL1-271 derivatives were synthesized and examined in a refined α-synuclein aggregation model as well as in an assay that monitors maturation of a disease-causing Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) mutant, which is also linked to Hsp70 function. Compared to the control, MAL1-271 significantly increased the number of cells lacking α-synuclein inclusions and increased the steady-state levels of the CFTR mutant. We also found that a nitrile-containing MAL1-271 analog exhibited similar effects in both assays. None of the derivatives exhibited cellular toxicity at concentrations up to 100 µm, nor were cellular stress response pathways induced. These data serve as a gateway for the continued development of a new class of Hsp70 agonists with efficacy in these and potentially other disease models.


Assuntos
Adenosina Trifosfatases/metabolismo , Ativadores de Enzimas/farmacologia , Ésteres/farmacologia , Proteínas de Choque Térmico HSP70/agonistas , Multimerização Proteica/efeitos dos fármacos , Pirimidinonas/farmacologia , Linhagem Celular Tumoral , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Ativadores de Enzimas/síntese química , Ativadores de Enzimas/química , Ativadores de Enzimas/toxicidade , Ésteres/síntese química , Ésteres/química , Ésteres/toxicidade , Células HEK293 , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Estrutura Molecular , Dobramento de Proteína/efeitos dos fármacos , Pirimidinonas/síntese química , Pirimidinonas/química , Pirimidinonas/toxicidade , Saccharomyces cerevisiae/enzimologia , Relação Estrutura-Atividade , alfa-Sinucleína/agonistas , alfa-Sinucleína/metabolismo
4.
Structure ; 25(12): 1839-1855.e11, 2017 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29153505

RESUMO

The discovery of ubistatins, small molecules that impair proteasomal degradation of proteins by directly binding to polyubiquitin, makes ubiquitin itself a potential therapeutic target. Although ubistatins have the potential for drug development and clinical applications, the lack of structural details of ubiquitin-ubistatin interactions has impeded their development. Here, we characterized a panel of new ubistatin derivatives using functional and binding assays. The structures of ubiquitin complexes with ubistatin B and hemi-ubistatin revealed direct interactions with ubiquitin's hydrophobic surface patch and the basic/polar residues surrounding it. Ubistatin B binds ubiquitin and diubiquitin tighter than a high-affinity ubiquitin receptor and shows strong preference for K48 linkages over K11 and K63. Furthermore, ubistatin B shields ubiquitin conjugates from disassembly by a range of deubiquitinases and by the 26S proteasome. Finally, ubistatin B penetrates cancer cells and alters the cellular ubiquitin landscape. These findings highlight versatile properties of ubistatins and have implications for their future development and use in targeting ubiquitin-signaling pathways.


Assuntos
Complexo de Endopeptidases do Proteassoma/química , Quinolinas/química , Ácidos Sulfanílicos/química , Ubiquitinas/química , Sítios de Ligação , Linhagem Celular , Células HeLa , Humanos , Simulação de Acoplamento Molecular , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Quinolinas/farmacologia , Saccharomyces cerevisiae/enzimologia , Ácidos Sulfanílicos/farmacologia , Ubiquitinas/antagonistas & inibidores , Ubiquitinas/metabolismo
5.
PLoS One ; 11(10): e0163615, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27732613

RESUMO

Small molecules that correct the folding defects and enhance surface localization of the F508del mutation in the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) comprise an important therapeutic strategy for cystic fibrosis lung disease. However, compounds that rescue the F508del mutant protein to wild type (WT) levels have not been identified. In this report, we consider obstacles to obtaining robust and therapeutically relevant levels of F508del CFTR. For example, markedly diminished steady state amounts of F508del CFTR compared to WT CFTR are present in recombinant bronchial epithelial cell lines, even when much higher levels of mutant transcript are present. In human primary airway cells, the paucity of Band B F508del is even more pronounced, although F508del and WT mRNA concentrations are comparable. Therefore, to augment levels of "repairable" F508del CFTR and identify small molecules that then correct this pool, we developed compound library screening protocols based on automated protein detection. First, cell-based imaging measurements were used to semi-quantitatively estimate distribution of F508del CFTR by high content analysis of two-dimensional images. We evaluated ~2,000 known bioactive compounds from the NIH Roadmap Molecular Libraries Small Molecule Repository in a pilot screen and identified agents that increase the F508del protein pool. Second, we analyzed ~10,000 compounds representing diverse chemical scaffolds for effects on total CFTR expression using a multi-plate fluorescence protocol and describe compounds that promote F508del maturation. Together, our findings demonstrate proof of principle that agents identified in this fashion can augment the level of endoplasmic reticulum (ER) resident "Band B" F508del CFTR suitable for pharmacologic correction. As further evidence in support of this strategy, PYR-41-a compound that inhibits the E1 ubiquitin activating enzyme-was shown to synergistically enhance F508del rescue by C18, a small molecule corrector. Our combined results indicate that increasing the levels of ER-localized CFTR available for repair provides a novel route to correct F508del CFTR.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/patologia , Retículo Endoplasmático/metabolismo , Bibliotecas de Moléculas Pequenas/química , Alelos , Benzoatos/química , Benzoatos/farmacologia , Células Cultivadas , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/química , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Furanos/química , Furanos/farmacologia , Deleção de Genes , Células HEK293 , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Ácidos Hidroxâmicos/química , Ácidos Hidroxâmicos/farmacologia , Microscopia de Fluorescência , Dobramento de Proteína , Estrutura Terciária de Proteína , Pirazóis/química , Pirazóis/farmacologia , RNA Mensageiro/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Ubiquitinação/efeitos dos fármacos , Vorinostat
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA