Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 295(23): 8017-8035, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32354745

RESUMEN

Niemann-Pick type C (NPC) disease is primarily caused by mutations in the NPC1 gene and is characterized by the accumulation of unesterified cholesterol and lipids in the late endosomal (LE) and lysosomal (Ly) compartments. The most prevalent disease-linked mutation is the I1061T variant of NPC1, which exhibits defective folding and trafficking from the endoplasmic reticulum to the LE/Ly compartments. We now show that the FDA-approved histone deacetylase inhibitor (HDACi) valproic acid (VPA) corrects the folding and trafficking defect associated with I1061T-NPC1 leading to restoration of cholesterol homeostasis, an effect that is largely driven by a reduction in HDAC7 expression. The VPA-mediated trafficking correction is in part associated with an increase in the acetylation of lysine residues in the cysteine-rich domain of NPC1. The HDACi-mediated correction is synergistically improved by combining it with the FDA-approved anti-malarial, chloroquine, a known lysosomotropic compound, which improved the stability of the LE/Ly-localized fraction of the I1061T variant. We posit that combining the activity of VPA, to modulate epigenetically the cellular acetylome, with chloroquine, to alter the lysosomal environment to favor stability of the trafficked I1061T variant protein can have a significant therapeutic benefit in patients carrying at least one copy of the I1061T variant of NPC1, the most common disease-associated mutation leading to NPC disease. Given its ability to cross the blood-brain barrier, we posit VPA provides a potential mechanism to improve the response to 2-hydroxypropyl-ß-cyclodextrin, by restoring a functional NPC1 to the cholesterol managing compartment as an adjunct therapy.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ácido Valproico/farmacología , Células Cultivadas , Cloroquina/farmacología , Colesterol/metabolismo , Células HeLa , Inhibidores de Histona Desacetilasas/química , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Estructura Molecular , Proteína Niemann-Pick C1 , Ácido Valproico/química
2.
Cell Chem Biol ; 27(3): 292-305.e6, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32017918

RESUMEN

Hsp90 plays an important role in health and is a therapeutic target for managing misfolding disease. Compounds that disrupt co-chaperone delivery of clients to Hsp90 target a subset of Hsp90 activities, thereby minimizing the toxicity of pan-Hsp90 inhibitors. Here, we have identified SEW04784 as a first-in-class inhibitor of the Aha1-stimulated Hsp90 ATPase activity without inhibiting basal Hsp90 ATPase. Nuclear magnetic resonance analysis reveals that SEW84 binds to the C-terminal domain of Aha1 to weaken its asymmetric binding to Hsp90. Consistent with this observation, SEW84 blocks Aha1-dependent Hsp90 chaperoning activities, including the in vitro and in vivo refolding of firefly luciferase, and the transcriptional activity of the androgen receptor in cell-based models of prostate cancer and promotes the clearance of phosphorylated tau in cellular and tissue models of neurodegenerative tauopathy. We propose that SEW84 provides a novel lead scaffold for developing therapeutic approaches to treat proteostatic disease.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Chaperonas Moleculares/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Células HEK293 , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Estructura Molecular , Pliegue de Proteína/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química
3.
Hum Mol Genet ; 29(1): 1-19, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31509197

RESUMEN

Genetic diversity provides a rich repository for understanding the role of proteostasis in the management of the protein fold in human biology. Failure in proteostasis can trigger multiple disease states, affecting both human health and lifespan. Niemann-Pick C1 (NPC1) disease is a rare genetic disorder triggered by mutations in NPC1, a multi-spanning transmembrane protein that is trafficked through the exocytic pathway to late endosomes (LE) and lysosomes (Ly) (LE/Ly) to globally manage cholesterol homeostasis. Defects triggered by >300 NPC1 variants found in the human population inhibit export of NPC1 protein from the endoplasmic reticulum (ER) and/or function in downstream LE/Ly, leading to cholesterol accumulation and onset of neurodegeneration in childhood. We now show that the allosteric inhibitor JG98, that targets the cytosolic Hsp70 chaperone/co-chaperone complex, can significantly improve the trafficking and post-ER protein level of diverse NPC1 variants. Using a new approach to model genetic diversity in human disease, referred to as variation spatial profiling, we show quantitatively how JG98 alters the Hsp70 chaperone/co-chaperone system to adjust the spatial covariance (SCV) tolerance and set-points on an amino acid residue-by-residue basis in NPC1 to differentially regulate variant trafficking, stability, and cholesterol homeostasis, results consistent with the role of BCL2-associated athanogene family co-chaperones in managing the folding status of NPC1 variants. We propose that targeting the cytosolic Hsp70 system by allosteric regulation of its chaperone/co-chaperone based client relationships can be used to adjust the SCV tolerance of proteostasis buffering capacity to provide an approach to mitigate systemic and neurological disease in the NPC1 population.


Asunto(s)
Variación Genética/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Proteína Niemann-Pick C1/metabolismo , Enfermedad de Niemann-Pick Tipo C/genética , Regulación Alostérica/genética , Regulación Alostérica/fisiología , Colesterol/metabolismo , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Variación Genética/genética , Proteínas HSP70 de Choque Térmico/genética , Humanos , Lisosomas/metabolismo , Proteína Niemann-Pick C1/genética
4.
J Cyst Fibros ; 19 Suppl 1: S33-S36, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31680043

RESUMEN

Mutations associated with cystic fibrosis (CF) have complex effects on the cystic fibrosis transmembrane conductance regulator (CFTR) protein. The most common CF mutation, F508del, disrupts the processing to and stability at the plasma membrane and function as a Cl- channel. CFTR is surrounded by a dynamic network of interacting components, referred to as the CFTR Functional Landscape, that impact its synthesis, folding, stability, trafficking and function. CFTR interacting proteins can be manipulated by functional genomic approaches to rescue the trafficking and functional defects characteristic of CF. Here we review recent efforts to elucidate the impact of genetic variation on the ability of the nascent CFTR polypeptide to interact with the proteostatic environment. We also provide an overview of how specific components of this protein network can be modulated to rescue the trafficking and functional defects associated with the F508del variant of CFTR. The identification of novel proteins playing key roles in the processing of CFTR could pave the way for their use as novel therapeutic targets to provide synergistic correction of mutant CFTR for the greater benefit of individuals with CF.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística , Terapia Genética/métodos , Transporte Iónico , Moduladores del Transporte de Membrana/farmacología , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Humanos , Transporte Iónico/efectos de los fármacos , Transporte Iónico/genética , Mutación , Proteostasis/efectos de los fármacos
5.
Nat Commun ; 10(1): 5052, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31699992

RESUMEN

To understand the impact of epigenetics on human misfolding disease, we apply Gaussian-process regression (GPR) based machine learning (ML) (GPR-ML) through variation spatial profiling (VSP). VSP generates population-based matrices describing the spatial covariance (SCV) relationships that link genetic diversity to fitness of the individual in response to histone deacetylases inhibitors (HDACi). Niemann-Pick C1 (NPC1) is a Mendelian disorder caused by >300 variants in the NPC1 gene that disrupt cholesterol homeostasis leading to the rapid onset and progression of neurodegenerative disease. We determine the sequence-to-function-to-structure relationships of the NPC1 polypeptide fold required for membrane trafficking and generation of a tunnel that mediates cholesterol flux in late endosomal/lysosomal (LE/Ly) compartments. HDACi treatment reveals unanticipated epigenomic plasticity in SCV relationships that restore NPC1 functionality. GPR-ML based matrices capture the epigenetic processes impacting information flow through central dogma, providing a framework for quantifying the effect of the environment on the healthspan of the individual.


Asunto(s)
Colesterol/metabolismo , Fibroblastos/metabolismo , Metabolismo de los Lípidos/genética , Proteína Niemann-Pick C1/genética , Enfermedad de Niemann-Pick Tipo C/genética , Línea Celular Tumoral , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Epigénesis Genética , Epigenómica , Fibroblastos/efectos de los fármacos , Células HeLa , Inhibidores de Histona Desacetilasas/farmacología , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Aprendizaje Automático , Proteína Niemann-Pick C1/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Distribución Normal , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/metabolismo , Análisis de Regresión , Relación Estructura-Actividad , Vorinostat/farmacología
6.
Hum Mol Genet ; 28(12): 1982-2000, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30753450

RESUMEN

Understanding the role of the epigenome in protein-misfolding diseases remains a challenge in light of genetic diversity found in the world-wide population revealed by human genome sequencing efforts and the highly variable response of the disease population to therapeutics. An ever-growing body of evidence has shown that histone deacetylase (HDAC) inhibitors (HDACi) can have significant benefit in correcting protein-misfolding diseases that occur in response to both familial and somatic mutation. Cystic fibrosis (CF) is a familial autosomal recessive disease, caused by genetic diversity in the CF transmembrane conductance regulator (CFTR) gene, a cyclic Adenosine MonoPhosphate (cAMP)-dependent chloride channel expressed at the apical plasma membrane of epithelial cells in multiple tissues. The potential utility of HDACi in correcting the phenylalanine 508 deletion (F508del) CFTR variant as well as the over 2000 CF-associated variants remains controversial. To address this concern, we examined the impact of US Food and Drug Administration-approved HDACi on the trafficking and function of a panel of CFTR variants. Our data reveal that panobinostat (LBH-589) and romidepsin (FK-228) provide functional correction of Class II and III CFTR variants, restoring cell surface chloride channel activity in primary human bronchial epithelial cells. We further demonstrate a synergistic effect of these HDACi with Vx809, which can significantly restore channel activity for multiple CFTR variants. These data suggest that HDACi can serve to level the cellular playing field for correcting CF-causing mutations, a leveling effect that might also extend to other protein-misfolding diseases.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Células Epiteliales/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Aminopiridinas/farmacología , Benzodioxoles/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/efectos de los fármacos , Inhibidores de Histona Desacetilasas/química , Humanos , Ácidos Hidroxámicos/farmacología , Mutación , Panobinostat/farmacología , Transporte de Proteínas/efectos de los fármacos , Eliminación de Secuencia , Sulfonamidas/farmacología
7.
J Biol Chem ; 293(35): 13682-13695, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-29986884

RESUMEN

The protein chaperones heat shock protein 70 (Hsp70) and Hsp90 are required for de novo folding of proteins and protect against misfolding-related cellular stresses by directing misfolded or slowly folding proteins to the ubiquitin/proteasome system (UPS) or autophagy/lysosomal degradation pathways. Here, we examined the role of the Bcl2-associated athanogene (BAG) family of Hsp70-specific nucleotide-exchange factors in the biogenesis and functional correction of genetic variants of the cystic fibrosis transmembrane conductance regulator (CFTR) whose mutations cause cystic fibrosis (CF). We show that siRNA-mediated silencing of BAG1 and -3, two BAG members linked to the clearance of misfolded proteins via the UPS and autophagy pathways, respectively, leads to functional correction of F508del-CFTR and other disease-associated CFTR variants. BAG3 silencing was the most effective, leading to improved F508del-CFTR stability, trafficking, and restoration of cell-surface function, both alone and in combination with the FDA-approved CFTR corrector, VX-809. We also found that the BAG3 silencing-mediated correction of F508del-CFTR restores the autophagy pathway, which is defective in F508del-CFTR-expressing cells, likely because of the maladaptive stress response in CF pathophysiology. These results highlight the potential therapeutic benefits of targeting the cellular chaperone system to improve the functional folding of CFTR variants contributing to CF and possibly other protein-misfolding-associated diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Mutación , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Estabilidad Proteica , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/genética , Regulación hacia Arriba
8.
J Biol Chem ; 293(35): 13477-13495, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30006345

RESUMEN

Inherited and somatic rare diseases result from >200,000 genetic variants leading to loss- or gain-of-toxic function, often caused by protein misfolding. Many of these misfolded variants fail to properly interact with other proteins. Understanding the link between factors mediating the transcription, translation, and protein folding of these disease-associated variants remains a major challenge in cell biology. Herein, we utilized the cystic fibrosis transmembrane conductance regulator (CFTR) protein as a model and performed a proteomics-based high-throughput screen (HTS) to identify pathways and components affecting the folding and function of the most common cystic fibrosis-associated mutation, the F508del variant of CFTR. Using a shortest-path algorithm we developed, we mapped HTS hits to the CFTR interactome to provide functional context to the targets and identified the eukaryotic translation initiation factor 3a (eIF3a) as a central hub for the biogenesis of CFTR. Of note, siRNA-mediated silencing of eIF3a reduced the polysome-to-monosome ratio in F508del-expressing cells, which, in turn, decreased the translation of CFTR variants, leading to increased CFTR stability, trafficking, and function at the cell surface. This finding suggested that eIF3a is involved in mediating the impact of genetic variations in CFTR on the folding of this protein. We posit that the number of ribosomes on a CFTR mRNA transcript is inversely correlated with the stability of the translated polypeptide. Polysome-based translation challenges the capacity of the proteostasis environment to balance message fidelity with protein folding, leading to disease. We suggest that this deficit can be corrected through control of translation initiation.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Factor 3 de Iniciación Eucariótica/metabolismo , Iniciación de la Cadena Peptídica Traduccional , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Factor 3 de Iniciación Eucariótica/genética , Humanos , Mutación , Fenilalanina/química , Fenilalanina/genética , Fenilalanina/metabolismo , Pliegue de Proteína , Mapas de Interacción de Proteínas , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/genética
9.
J Mol Biol ; 430(18 Pt A): 2951-2973, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-29924966

RESUMEN

The advent of precision medicine for genetic diseases has been hampered by the large number of variants that cause familial and somatic disease, a complexity that is further confounded by the impact of genetic modifiers. To begin to understand differences in onset, progression and therapeutic response that exist among disease-causing variants, we present the proteomic variant approach (ProVarA), a proteomic method that integrates mass spectrometry with genomic tools to dissect the etiology of disease. To illustrate its value, we examined the impact of variation in cystic fibrosis (CF), where 2025 disease-associated mutations in the CF transmembrane conductance regulator (CFTR) gene have been annotated and where individual genotypes exhibit phenotypic heterogeneity and response to therapeutic intervention. A comparative analysis of variant-specific proteomics allows us to identify a number of protein interactions contributing to the basic defects associated with F508del- and G551D-CFTR, two of the most common disease-associated variants in the patient population. We demonstrate that a number of these causal interactions are significantly altered in response to treatment with Vx809 and Vx770, small-molecule therapeutics that respectively target the F508del and G551D variants. ProVarA represents the first comparative proteomic analysis among multiple disease-causing mutations, thereby providing a methodological approach that provides a significant advancement to existing proteomic efforts in understanding the impact of variation in CF disease. We posit that the implementation of ProVarA for any familial or somatic mutation will provide a substantial increase in the knowledge base needed to implement a precision medicine-based approach for clinical management of disease.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Medicina de Precisión , Proteómica , Biomarcadores , Fibrosis Quística/diagnóstico , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Perfilación de la Expresión Génica , Estudios de Asociación Genética/métodos , Variación Genética , Genotipo , Humanos , Mutación , Medicina de Precisión/métodos , Proteoma , Proteómica/métodos
10.
J Lipid Res ; 58(4): 695-708, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28193631

RESUMEN

Niemann-Pick C (NPC) disease is an autosomal recessive disorder that leads to excessive storage of cholesterol and other lipids in late endosomes and lysosomes. The large majority of NPC disease is caused by mutations in NPC1, a large polytopic membrane protein that functions in late endosomes. There are many disease-associated mutations in NPC1, and most patients are compound heterozygotes. The most common mutation, NPC1I1061T, has been shown to cause endoplasmic reticulum-associated degradation of the NPC1 protein. Treatment of patient-derived NPC1I1061T fibroblasts with histone deacetylase inhibitors (HDACis) vorinostat or panobinostat increases expression of the mutant NPC1 protein and leads to correction of the cholesterol storage. Here, we show that several other human NPC1 mutant fibroblast cell lines can also be corrected by vorinostat or panobinostat and that treatment with vorinostat extends the lifetime of the NPC1I1061T protein. To test effects of HDACi on a large number of NPC1 mutants, we engineered a U2OS cell line to suppress NPC1 expression by shRNA and then transiently transfected these cells with 60 different NPC1 mutant constructs. The mutant NPC1 did not significantly reduce cholesterol accumulation, but approximately 85% of the mutants showed reduced cholesterol accumulation when treated with vorinostat or panobinostat.


Asunto(s)
Proteínas Portadoras/genética , Colesterol/metabolismo , Inhibidores de Histona Desacetilasas/administración & dosificación , Glicoproteínas de Membrana/genética , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Proteínas Portadoras/antagonistas & inhibidores , Línea Celular , Degradación Asociada con el Retículo Endoplásmico/efectos de los fármacos , Endosomas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Ácidos Hidroxámicos/administración & dosificación , Indoles/administración & dosificación , Péptidos y Proteínas de Señalización Intracelular , Lisosomas/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Mutación , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/metabolismo , Enfermedad de Niemann-Pick Tipo C/patología , Panobinostat , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transfección , Vorinostat
11.
Methods Mol Biol ; 1510: 77-91, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27761814

RESUMEN

Gene expression is regulated in part through the reversible acetylation of histones, by the action of histone acetyltransferases (HAT) and histone deacetylases (HDAC). HAT activity results in the addition of acetyl groups on the lysine residues of histone tails leading to decondensation of the chromatin, and increased gene transcription in general, whereas HDACs remove these acetyl groups, thus leading to an overall suppression of gene transcription. Recent evidence has elucidated that histones are not the only components of the proteome that are targeted by HATs and HDACs. A large number of nonhistone proteins undergo posttranslational acetylation. They include proteins involved in mRNA stability, protein localization and degradation, as well as protein-protein and protein-DNA interactions. In recent years, numerous studies have discovered increased HDAC expression and/or activity in numerous disease states, including cancer, where the upregulation of HDAC family members leads to dysregulation of genes and proteins involved in cell proliferation, cell cycle regulation, and apoptosis. These observations have pushed HDAC inhibitors (HDACi) to the forefront of therapeutic development of oncological conditions. HDACi, such as Vorinostat (Suberoylanilide hydroxamic acid (SAHA)), affect cancer cells in part by suppressing the translation of key proteins linked to tumorigenesis, such as cyclin D1 and hypoxia inducible factor 1 alpha (HIF-1α). Herein we describe methodologies to analyze the impact of the HDACi Vorinostat on HIF-1α translational regulation and downstream effectors.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histonas/genética , Ácidos Hidroxámicos/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Acetilación , Western Blotting/métodos , Línea Celular Tumoral , Cromatina/química , Cromatina/efectos de los fármacos , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Deferoxamina/farmacología , Factor 3 de Iniciación Eucariótica/antagonistas & inhibidores , Factor 3 de Iniciación Eucariótica/genética , Factor 3 de Iniciación Eucariótica/metabolismo , Regulación Neoplásica de la Expresión Génica , Glicina/análogos & derivados , Glicina/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Leupeptinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Vorinostat
12.
PLoS Biol ; 12(11): e1001998, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25406061

RESUMEN

Diseases of protein folding arise because of the inability of an altered peptide sequence to properly engage protein homeostasis components that direct protein folding and function. To identify global principles of misfolding disease pathology we examined the impact of the local folding environment in alpha-1-antitrypsin deficiency (AATD), Niemann-Pick type C1 disease (NPC1), Alzheimer's disease (AD), and cystic fibrosis (CF). Using distinct models, including patient-derived cell lines and primary epithelium, mouse brain tissue, and Caenorhabditis elegans, we found that chronic expression of misfolded proteins not only triggers the sustained activation of the heat shock response (HSR) pathway, but that this sustained activation is maladaptive. In diseased cells, maladaptation alters protein structure-function relationships, impacts protein folding in the cytosol, and further exacerbates the disease state. We show that down-regulation of this maladaptive stress response (MSR), through silencing of HSF1, the master regulator of the HSR, restores cellular protein folding and improves the disease phenotype. We propose that restoration of a more physiological proteostatic environment will strongly impact the management and progression of loss-of-function and gain-of-toxic-function phenotypes common in human disease.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/etiología , Proteínas de Unión al ADN/genética , Deficiencias en la Proteostasis/genética , Factores de Transcripción/genética , Animales , Antineoplásicos Alquilantes/uso terapéutico , Caenorhabditis elegans , Línea Celular , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/metabolismo , Proteínas de Unión al ADN/metabolismo , Diterpenos/uso terapéutico , Evaluación Preclínica de Medicamentos , Compuestos Epoxi/uso terapéutico , Silenciador del Gen , Factores de Transcripción del Choque Térmico , Humanos , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Ratones Transgénicos , Organoides , Fenantrenos/uso terapéutico , Prostaglandina-E Sintasas , Pliegue de Proteína , Mucosa Respiratoria/metabolismo , Estrés Fisiológico , Factores de Transcripción/metabolismo
13.
PLoS One ; 9(8): e106224, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25166596

RESUMEN

Hypoxia inducible factor 1α (HIF-1α) is a master regulator of tumor angiogenesis being one of the major targets for cancer therapy. Previous studies have shown that Histone Deacetylase Inhibitors (HDACi) block tumor angiogenesis through the inhibition of HIF-1α expression. As such, Vorinostat (Suberoylanilide Hydroxamic Acid/SAHA) and Romidepsin, two HDACis, were recently approved by the Food and Drug Administration (FDA) for the treatment of cutaneous T cell lymphoma. Although HDACis have been shown to affect HIF-1α expression by modulating its interactions with the Hsp70/Hsp90 chaperone axis or its acetylation status, the molecular mechanisms by which HDACis inhibit HIF-1α expression need to be further characterized. Here, we report that the FDA-approved HDACi Vorinostat/SAHA inhibits HIF-1α expression in liver cancer-derived cell lines, by a new mechanism independent of p53, prolyl-hydroxylases, autophagy and proteasome degradation. We found that SAHA or silencing of HDAC9 mechanism of action is due to inhibition of HIF-1α translation, which in turn, is mediated by the eukaryotic translation initiation factor--eIF3G. We also highlighted that HIF-1α translation is dramatically inhibited when SAHA is combined with eIF3H silencing. Taken together, we show that HDAC activity regulates HIF-1α translation, with HDACis such as SAHA representing a potential novel approach for the treatment of hepatocellular carcinoma.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Hepáticas/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Línea Celular Tumoral , Factor 3 de Iniciación Eucariótica/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Histona Desacetilasas , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Vorinostat
14.
Artículo en Inglés | MEDLINE | ID: mdl-23426524

RESUMEN

The folding biology common to all three kingdoms of life (Archaea, Bacteria, and Eukarya) is proteostasis. The proteostasis network (PN) functions as a "cloud" to generate, protect, and degrade the proteome. Whereas microbes (Bacteria, Archaea) have a single compartment, Eukarya have numerous subcellular compartments. We examine evidence that Eukarya compartments use coat, tether, and fusion (CTF) membrane trafficking components to form an evolutionarily advanced arm of the PN that we refer to as the "trafficking PN" (TPN). We suggest that the TPN builds compartments by generating a mosaic of integrated cargo-specific trafficking signatures (TRaCKS). TRaCKS control the temporal and spatial features of protein-folding biology based on the Anfinsen principle that the local environment plays a critical role in managing protein structure. TPN-generated endomembrane compartments apply a "quinary" level of structural control to modify the secondary, tertiary, and quaternary structures defined by the primary polypeptide-chain sequence. The development of Anfinsen compartments provides a unifying foundation for understanding the purpose of endomembrane biology and its capacity to drive extant Eukarya function and diversity.


Asunto(s)
Membrana Celular/fisiología , Eucariontes/metabolismo , Modelos Biológicos , Transporte de Proteínas/fisiología , Proteoma , Membrana Celular/química , Proteínas de Transporte de Membrana/fisiología
15.
J Biol Chem ; 287(45): 38265-78, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22995909

RESUMEN

α1-Antitrypsin (α1AT) deficiency (α1ATD) is a consequence of defective folding, trafficking, and secretion of α1AT in response to a defect in its interaction with the endoplasmic reticulum proteostasis machineries. The most common and severe form of α1ATD is caused by the Z-variant and is characterized by the accumulation of α1AT polymers in the endoplasmic reticulum of the liver leading to a severe reduction (>85%) of α1AT in the serum and its anti-protease activity in the lung. In this organ α1AT is critical for ensuring tissue integrity by inhibiting neutrophil elastase, a protease that degrades elastin. Given the limited therapeutic options in α1ATD, a more detailed understanding of the folding and trafficking biology governing α1AT biogenesis and its response to small molecule regulators is required. Herein we report the correction of Z-α1AT secretion in response to treatment with the histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA), acting in part through HDAC7 silencing and involving a calnexin-sensitive mechanism. SAHA-mediated correction restores Z-α1AT secretion and serpin activity to a level 50% that observed for wild-type α1AT. These data suggest that HDAC activity can influence Z-α1AT protein traffic and that SAHA may represent a potential therapeutic approach for α1ATD and other protein misfolding diseases.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Deficiencia de alfa 1-Antitripsina/prevención & control , alfa 1-Antitripsina/metabolismo , Calnexina/genética , Calnexina/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/metabolismo , Expresión Génica/efectos de los fármacos , Células HCT116 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Immunoblotting , Hígado/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutación , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Pliegue de Proteína , Transporte de Proteínas/efectos de los fármacos , Deficiencias en la Proteostasis/sangre , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/prevención & control , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vorinostat , alfa 1-Antitripsina/sangre , alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/sangre , Deficiencia de alfa 1-Antitripsina/genética
16.
PLoS One ; 7(5): e37682, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22701530

RESUMEN

Protein folding is the primary role of proteostasis network (PN) where chaperone interactions with client proteins determine the success or failure of the folding reaction in the cell. We now address how the Phe508 deletion in the NBD1 domain of the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) protein responsible for cystic fibrosis (CF) impacts the binding of CFTR with cellular chaperones. We applied single ion reaction monitoring mass spectrometry (SRM-MS) to quantitatively characterize the stoichiometry of the heat shock proteins (Hsps) in CFTR folding intermediates in vivo and mapped the sites of interaction of the NBD1 domain of CFTR with Hsp90 in vitro. Unlike folding of WT-CFTR, we now demonstrate the presence of ΔF508-CFTR in a stalled folding intermediate in stoichiometric association with the core Hsps 40, 70 and 90, referred to as a 'chaperone trap'. Culturing cells at 30 C resulted in correction of ΔF508-CFTR trafficking and function, restoring the sub-stoichiometric association of core Hsps observed for WT-CFTR. These results support the interpretation that ΔF508-CFTR is restricted to a chaperone-bound folding intermediate, a state that may contribute to its loss of trafficking and increased targeting for degradation. We propose that stalled folding intermediates could define a critical proteostasis pathway branch-point(s) responsible for the loss of function in misfolding diseases as observed in CF.


Asunto(s)
Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Espectrometría de Masas , Pliegue de Proteína
17.
J Biol Chem ; 287(26): 21914-25, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22474283

RESUMEN

Cystic fibrosis (CF) is caused by mutations in the apical chloride channel cystic fibrosis transmembrane conductance regulator (CFTR) with 90% of patients carrying at least one deletion of the F508 (ΔF508) allele. This mutant form of CFTR is characterized by a folding and trafficking defect that prevents exit from the endoplasmic reticulum. We previously reported that ΔF508 CFTR can be recovered in a complex with Hsp90 and its co-chaperones as an on-pathway folding intermediate, suggesting that Δ508 CF disease arises due to a failure of the proteostasis network (PN), which manages protein folding and degradation in the cell. We have now examined the role of FK506-binding protein 8 (FKBP8), a component of the CFTR interactome, during the biogenesis of wild-type and ΔF508 CFTR. FKBP8 is a member of the peptidylprolyl isomerase family that mediates the cis/trans interconversion of peptidyl prolyl bonds. Our results suggest that FKBP8 is a key PN factor required at a post-Hsp90 step in CFTR biogenesis. In addition, changes in its expression level or alteration of its activity by a peptidylprolyl isomerase inhibitor alter CFTR stability and transport. We propose that CF is caused by the sequential failure of the prevailing PN pathway to stabilize ΔF508-CFTR for endoplasmic reticulum export, a pathway that can be therapeutically managed.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Proteínas de Unión a Tacrolimus/química , ADN/química , Retículo Endoplásmico/metabolismo , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Proteínas de Choque Térmico/metabolismo , Humanos , Inmunofilinas/metabolismo , Yoduros/química , Desnaturalización Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Temperatura , Factores de Tiempo
18.
Nat Chem Biol ; 8(2): 185-96, 2011 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-22198733

RESUMEN

Protein homeostasis (proteostasis) is essential for cellular and organismal health. Stress, aging and the chronic expression of misfolded proteins, however, challenge the proteostasis machinery and the vitality of the cell. Enhanced expression of molecular chaperones, regulated by heat shock transcription factor-1 (HSF-1), has been shown to restore proteostasis in a variety of conformational disease models, suggesting this mechanism as a promising therapeutic approach. We describe the results of a screen comprised of ∼900,000 small molecules that identified new classes of small-molecule proteostasis regulators that induce HSF-1-dependent chaperone expression and restore protein folding in multiple conformational disease models. These beneficial effects to proteome stability are mediated by HSF-1, FOXO, Nrf-2 and the chaperone machinery through mechanisms that are distinct from current known small-molecule activators of the heat shock response. We suggest that modulation of the proteostasis network by proteostasis regulators may be a promising therapeutic approach for the treatment of a variety of protein conformational diseases.


Asunto(s)
Evaluación Preclínica de Medicamentos , Chaperonas Moleculares/efectos de los fármacos , Proteínas/efectos de los fármacos , Deficiencias en la Proteostasis/tratamiento farmacológico , Factores de Transcripción/efectos de los fármacos , Animales , Caenorhabditis elegans , Línea Celular , Proteínas de Unión al ADN/efectos de los fármacos , Factores de Transcripción Forkhead/efectos de los fármacos , Factores de Transcripción del Choque Térmico , Homeostasis/efectos de los fármacos , Humanos , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Proteínas/química , Proteínas/fisiología , Ratas
19.
ACS Med Chem Lett ; 2(9): 703-707, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21984958

RESUMEN

Cystic fibrosis (CF) is a loss-of-function disease caused by mutations in the CF transmembrane conductance regulator (CFTR) protein, a chloride ion channel that localizes to the apical plasma membrane of epithelial cells. The most common form of the disease results from the deletion of phenylalanine-508 (ΔF508), leading to the accumulation of CFTR in the endoplasmic reticulum with a concomitant loss of chloride flux. We discovered that cyclic tetrapeptides, such as 11, 14, and 15, are able to correct the trafficking defect and restore cell surface activity of ΔF508-CFTR. Although this class of cyclic tetrapeptides is known to contain inhibitors of certain histone deacetylase (HDAC) isoforms, their HDAC inhibitory potencies did not directly correlate with their ability to rescue ΔF508-CFTR. In full HDAC profiling, 15 strongly inhibited HDACs 1, 2, 3, 10 and 11, but not HDACs 4-9. Although 15 had less potent IC(50) values than reference agent vorinostat (2) in HDAC profiling, it was markedly more potent than 2 in rescuing ΔF508-CFTR. We suggest that specific HDACs can have a differential influence on correcting ΔF508-CFTR, which may reflect both deacetylase and protein scaffolding actions.

20.
Artículo en Inglés | MEDLINE | ID: mdl-21421917

RESUMEN

Cystic fibrosis (CF) is a consequence of defective recognition of the multimembrane spanning protein cystic fibrosis conductance transmembrane regulator (CFTR) by the protein homeostasis or proteostasis network (PN) (Hutt and Balch (2010). Like many variant proteins triggering misfolding diseases, mutant CFTR has a complex folding and membrane trafficking itinerary that is managed by the PN to maintain proteome balance and this balance is disrupted in human disease. The biological pathways dictating the folding and function of CFTR in health and disease are being studied by numerous investigators, providing a unique opportunity to begin to understand and therapeutically address the role of the PN in disease onset, and its progression during aging. We discuss the general concept that therapeutic management of the emergent properties of the PN to control the energetics of CFTR folding biology may provide significant clinical benefit.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Descubrimiento de Drogas/métodos , Pliegue de Proteína , Deficiencias en la Proteostasis/metabolismo , Fibrosis Quística/tratamiento farmacológico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Manejo de la Enfermedad , Humanos , Modelos Biológicos , Mutación/genética , Transporte de Proteínas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...