Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
ACS Chem Biol ; 18(5): 1124-1135, 2023 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-37144894

RESUMEN

The accumulation and aggregation of the microtubule-associated protein tau (tau) into intracellular neuronal tangles are a hallmark of a range of progressive neurodegenerative tauopathies, including Alzheimer's disease (AD), frontotemporal dementia, Pick's disease, and progressive supranuclear palsy. The aberrant phosphorylation of tau is associated with tau aggregates in AD. Members of the heat shock protein 70 kDa (Hsp70) family of chaperones bind directly to tau and modulate tau clearance and aggregation. Small molecules that inhibit the Hsp70 family of chaperones have been shown to reduce the accumulation of tau, including phosphorylated tau. Here, eight analogs of the rhodacyanine inhibitor, JG-98, were synthesized and evaluated. Like JG-98, many of the compounds inhibited ATPase activity of the cytosolic heat shock cognate 70 protein (Hsc70) and reduced total, aggregated, and phosphorylated tau accumulation in cultured cells. Three compounds, representing divergent clogP values, were evaluated for in vivo blood-brain barrier penetration and tau reduction in an ex vivo brain slice model. AL69, the compound with the lowest clogP and the lowest membrane retention in a parallel artificial membrane permeability assay (PAMPA), reduced phosphorylated tau accumulation. Our results suggest that benzothiazole substitutions of JG-98 that increase hydrophilicity may increase the efficacy of these Hsp70 inhibitors to reduce phosphorylated tau.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Benzotiazoles/farmacología , Proteínas HSP70 de Choque Térmico , Chaperonas Moleculares , Proteínas tau/metabolismo , Tauopatías/metabolismo
2.
Protein Sci ; 31(11): e4448, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36305768

RESUMEN

Tauopathies, such as Alzheimer's disease, are characterized by the misfolding and progressive accumulation of the microtubule associated protein tau. Chaperones, tasked with maintaining protein homeostasis, can become imbalanced with age and contribute to the progression of neurodegenerative disease. Cyclophilins are a promising pool of underinvestigated chaperones with peptidyl-prolyl isomerase activity that may play protective roles in regulating tau aggregation. Using a Thioflavin T fluorescence-based assay to monitor in vitro tau aggregation, all eight cyclophilins, which include PPIA to PPIH prevent tau aggregation, with PPIB, PPIC, PPID, and PPIH showing the greatest inhibition. The low thermal stability of PPID and the strong heparin binding of PPIB undermines the simplistic interpretation of reduced tau aggregation. In a cellular model of tau accumulation, all cyclophilins, except PPID and PPIH, reduce insoluble tau. PPIB, PPIC, PPIE, and PPIF also reduce soluble tau levels with PPIC exclusively protecting cells from tau seeding. Overall, this study demonstrates cyclophilins prevent tau fibril formation and many reduce cellular insoluble tau accumulation with PPIC having the greatest potential as a molecular tool to mitigate tau seeding and accumulation.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Humanos , Ciclofilinas/química , Ciclofilinas/metabolismo , Proteínas tau/metabolismo , Pliegue de Proteína , Chaperonas Moleculares/metabolismo , Enfermedad de Alzheimer/metabolismo
3.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055033

RESUMEN

The microtubule-associated protein tau pathologically accumulates and aggregates in Alzheimer's disease (AD) and other tauopathies, leading to cognitive dysfunction and neuronal loss. Molecular chaperones, like small heat-shock proteins (sHsps), can help deter the accumulation of misfolded proteins, such as tau. Here, we tested the hypothesis that the overexpression of wild-type Hsp22 (wtHsp22) and its phosphomimetic (S24,57D) Hsp22 mutant (mtHsp22) could slow tau accumulation and preserve memory in a murine model of tauopathy, rTg4510. Our results show that Hsp22 protected against deficits in synaptic plasticity and cognition in the tauopathic brain. However, we did not detect a significant change in tau phosphorylation or levels in these mice. This led us to hypothesize that the functional benefit was realized through the restoration of dysfunctional pathways in hippocampi of tau transgenic mice since no significant benefit was measured in non-transgenic mice expressing wtHsp22 or mtHsp22. To identify these pathways, we performed mass spectrometry of tissue lysates from the injection site. Overall, our data reveal that Hsp22 overexpression in neurons promotes synaptic plasticity by regulating canonical pathways and upstream regulators that have been characterized as potential AD markers and synaptogenesis regulators, like EIF4E and NFKBIA.


Asunto(s)
Encéfalo/metabolismo , Cognición , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Aprendizaje , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Tauopatías/etiología , Tauopatías/metabolismo , Animales , Biomarcadores , Encéfalo/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Metabolismo Energético , Expresión Génica , Ratones , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Fosforilación , Transducción de Señal , Tauopatías/patología , Transducción Genética , Proteínas tau/genética , Proteínas tau/metabolismo
4.
NPJ Aging Mech Dis ; 7(1): 9, 2021 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-33941782

RESUMEN

Abnormal accumulation of hyperphosphorylated tau induces pathogenesis in neurodegenerative diseases, like Alzheimer's disease. Molecular chaperones with peptidyl-prolyl cis/trans isomerase (PPIase) activity are known to regulate these processes. Previously, in vitro studies have shown that the 52 kDa FK506-binding protein (FKBP52) interacts with tau inducing its oligomerization and fibril formation to promote toxicity. Thus, we hypothesized that increased expression of FKBP52 in the brains of tau transgenic mice would alter tau phosphorylation and neurofibrillary tangle formation ultimately leading to memory impairments. To test this, tau transgenic (rTg4510) and wild-type mice received bilateral hippocampal injections of virus overexpressing FKBP52 or GFP control. We examined hippocampal-dependent memory, synaptic plasticity, tau phosphorylation status, and neuronal health. This work revealed that rTg4510 mice overexpressing FKBP52 had impaired spatial learning, accompanied by long-term potentiation deficits and hippocampal neuronal loss, which was associated with a modest increase in total caspase 12. Together with previous studies, our findings suggest that FKBP52 may sensitize neurons to tau-mediated dysfunction via activation of a caspase-dependent pathway, contributing to memory and learning impairments.

5.
Protein Sci ; 30(7): 1350-1359, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33686711

RESUMEN

Alzheimer's disease is a progressive fatal neurodegenerative disease with no cure or effective treatments. The hallmarks of disease include extracellular plaques and intracellular tangles of aggregated protein. The intracellular tangles consist of the microtubule associated protein tau. Preventing the pathological aggregation of tau may be an important therapeutic approach to treat disease. In this study we show that small heat shock protein 22 kDa (Hsp22) can prevent the aggregation of tau in vitro. Additionally, tau can undergo liquid-liquid phase separation (LLPS) in the presence of crowding reagents which causes it to have an increased aggregation rate. We show that Hsp22 can modulate both the aggregation and LLPS behavior of tau in vitro.


Asunto(s)
Proteínas de Choque Térmico/química , Chaperonas Moleculares/química , Agregado de Proteínas , Proteínas tau/química , Humanos
6.
J Clin Invest ; 131(4)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33586680

RESUMEN

Tauopathies display a spectrum of phenotypes from cognitive to affective behavioral impairments; however, mechanisms promoting tau pathology and how tau elicits behavioral impairment remain unclear. We report a unique interaction between polyamine metabolism, behavioral impairment, and tau fate. Polyamines are ubiquitous aliphatic molecules that support neuronal function, axonal integrity, and cognitive processing. Transient increases in polyamine metabolism hallmark the cell's response to various insults, known as the polyamine stress response (PSR). Dysregulation of gene transcripts associated with polyamine metabolism in Alzheimer's disease (AD) brains were observed, and we found that ornithine decarboxylase antizyme inhibitor 2 (AZIN2) increased to the greatest extent. We showed that sustained AZIN2 overexpression elicited a maladaptive PSR in mice with underlying tauopathy (MAPT P301S; PS19). AZIN2 also increased acetylpolyamines, augmented tau deposition, and promoted cognitive and affective behavioral impairments. Higher-order polyamines displaced microtubule-associated tau to facilitate polymerization but also decreased tau seeding and oligomerization. Conversely, acetylpolyamines promoted tau seeding and oligomers. These data suggest that tauopathies launch an altered enzymatic signature that endorses a feed-forward cycle of disease progression. Taken together, the tau-induced PSR affects behavior and disease continuance, but may also position the polyamine pathway as a potential entry point for plausible targets and treatments of tauopathy, including AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Poliaminas Biogénicas/metabolismo , Carboxiliasas/metabolismo , Proteínas Portadoras/metabolismo , Hipocampo/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Carboxiliasas/genética , Proteínas Portadoras/genética , Femenino , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteínas tau/genética , Proteínas tau/metabolismo
7.
Int J Mol Sci ; 21(15)2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32751642

RESUMEN

Misfolding, aggregation and accumulation of proteins are toxic elements in the progression of a broad range of neurodegenerative diseases. Molecular chaperones enable a cellular defense by reducing or compartmentalizing these insults. Small heat shock proteins (sHsps) engage proteins early in the process of misfolding and can facilitate their proper folding or refolding, sequestration, or clearance. Here, we evaluate the effects of the sHsp Hsp22, as well as a pseudophosphorylated mutant and an N-terminal domain deletion (NTDΔ) variant on tau aggregation in vitro and tau accumulation and aggregation in cultured cells. Hsp22 wild-type (WT) protein had a significant inhibitory effect on heparin-induced aggregation in vitro and the pseudophosphorylated mutant Hsp22 demonstrated a similar effect. When co-expressed in a cell culture model with tau, these Hsp22 constructs significantly reduced soluble tau protein levels when transfected at a high ratio relative to tau. However, the Hsp22 NTDΔ protein drastically reduced the soluble protein expression levels of both tau WT and tau P301L/S320F even at lower transfection ratios, which resulted in a correlative reduction of the triton-insoluble tau P301L/S320F aggregates.


Asunto(s)
Proteínas de Choque Térmico/genética , Chaperonas Moleculares/genética , Enfermedades Neurodegenerativas/genética , Proteínas tau/genética , Animales , Regulación de la Expresión Génica/genética , Proteínas de Choque Térmico Pequeñas/genética , Humanos , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/patología , Agregación Patológica de Proteínas/genética , Unión Proteica/genética , Deficiencias en la Proteostasis/genética
8.
Cells ; 9(1)2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31947657

RESUMEN

Tau dysfunction is common in several neurodegenerative diseases including Alzheimer's disease (AD) and frontotemporal dementia (FTD). Affective symptoms have often been associated with aberrant tau pathology and are commonly comorbid in patients with tauopathies, indicating a connection between tau functioning and mechanisms of depression. The current study investigated depression-like behavior in Mapt-/- mice, which contain a targeted deletion of the gene coding for tau. We show that 6-month Mapt-/- mice are resistant to depressive behaviors, as evidenced by decreased immobility time in the forced swim and tail suspension tests, as well as increased escape behavior in a learned helplessness task. Since depression has also been linked to deficient adult neurogenesis, we measured neurogenesis in the hippocampal dentate gyrus and subventricular zone using 5-bromo-2-deoxyuridine (BrdU) labeling. We found that neurogenesis is increased in the dentate gyrus of 14-month-old Mapt-/- brains compared to wild type, providing a potential mechanism for their behavioral phenotypes. In addition to the hippocampus, an upregulation of proteins involved in neurogenesis was observed in the frontal cortex and amygdala of the Mapt-/- mice using proteomic mass spectrometry. All together, these findings suggest that tau may have a role in the depressive symptoms observed in many neurodegenerative diseases and identify tau as a potential molecular target for treating depression.


Asunto(s)
Hipocampo/metabolismo , Neurogénesis , Neuronas/metabolismo , Proteínas tau/deficiencia , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología
9.
Brain Behav Immun Health ; 9: 100143, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34589890

RESUMEN

Early life stress (ELS) adversely affects the brain and is commonly associated with the etiology of mental health disorders, like depression. In addition to the mood-related symptoms, patients with depression show dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, increased peripheral inflammation, and structural brain alterations. Although the underlying causes are unknown, polymorphisms in the FK506-binding protein 5 (FKBP5) gene, a regulator of glucocorticoid receptor (GR) activity, interact with childhood adversities to increase vulnerability to depressive disorders. We hypothesized that high FKBP5 protein levels combined with early life stress (ELS) would alter the HPA axis and brain, promoting depressive-like behaviors. To test this, we exposed males and females of a mouse model overexpressing FKBP5 in the brain (rTgFKBP5 mice), or littermate controls, to maternal separation for 14 days after birth. Then, we evaluated neuroendocrine, behavioral, and brain changes in young adult and aged mice. We observed lower basal corticosterone (CORT) levels in rTgFKBP5 mice, which was exacerbated in females. Aged, but not young, rTgFKBP5 mice showed increased depressive-like behaviors. Moreover, FKBP5 overexpression reduced hippocampal neuron density in aged mice, while promoting markers of microglia expression, but these effects were reversed by ELS. Together, these results demonstrate that high FKBP5 affects basal CORT levels, depressive-like symptoms, and numbers of neurons and microglia in the hippocampus in an age-dependent manner.

10.
J Biol Chem ; 294(34): 12717-12728, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31270212

RESUMEN

Nonsynonymous gene mutations can be beneficial, neutral, or detrimental to the stability, structure, and biological function of the encoded protein, but the effects of these mutations are often not readily predictable. For example, the ß-propeller olfactomedin domain of myocilin (mOLF) exhibits a complex interrelationship among structure(s), stability, and aggregation. Numerous mutations within mOLF are linked to glaucoma; the resulting variants are less stable, aggregation-prone, and sequestered intracellularly, causing cytotoxicity. Here, we report the first stable mOLF variants carrying substitutions in the calcium-binding site that exhibit solution characteristics indistinguishable from those of glaucoma variants. Crystal structures of these stable variants at 1.8-2.0-Å resolution revealed features that we could not predict by molecular dynamics simulations, including loss of loop structure, helix unwinding, and a blade shift. Double mutants that combined a stabilizing substitution and a selected glaucoma-causing single-point mutant rescued in vitro folding and stability defects. In the context of full-length myocilin, secretion of stable single variants was indistinguishable from that of the WT protein, and the double mutants were secreted to varying extents. In summary, our finding that mOLF can tolerate particular substitutions that render the protein stable despite a conformational switch emphasizes the complexities in differentiating between benign and glaucoma-causing variants and provides new insight into the possible biological function of myocilin.


Asunto(s)
Proteínas del Citoesqueleto/genética , Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo/genética , Glaucoma/genética , Glicoproteínas/genética , Mutación , Proteínas del Citoesqueleto/química , Proteínas de la Matriz Extracelular/química , Proteínas del Ojo/química , Variación Genética/genética , Glicoproteínas/química , Células HEK293 , Humanos , Simulación de Dinámica Molecular
11.
Int J Mol Sci ; 20(11)2019 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167373

RESUMEN

Clinical studies show a significant association of childhood adversities and FK506-binding protein 5 (FKBP5) polymorphisms on increasing the susceptibility for neuropsychiatric disorders. However, the mechanisms by which early life stress (ELS) influences FKBP5 actions have not been fully elucidated. We hypothesized that interactions between ELS and high FKBP5 induce phenotypic changes that correspond to underlying molecular changes in the brain. To test this, we exposed newborn mice overexpressing human FKBP5 in the forebrain, rTgFKBP5, to ELS using a maternal separation. Two months after ELS, we observed that ELS increased anxiety levels, specifically in mice overexpressing FKBP5, an effect that was more pronounced in females. Biochemically, Protein kinase B (AKT) phosphorylation was reduced in the dorsal hippocampus in rTgFKBP5 mice, which demonstrates that significant molecular changes occur as a result of ELS when FKBP5 levels are altered. Taken together, our results have a significant impact on our understanding mechanisms underlying the gene x environment interaction showing that anxiety and AKT signaling in the hippocampus were affected by the combination of ELS and FKBP5. An increased knowledge of the molecular mechanisms underlying these interactions may help determine if FKBP5 could be an effective target for the treatment of anxiety and other mood-related illnesses.


Asunto(s)
Trastornos de Ansiedad/etiología , Trastornos de Ansiedad/metabolismo , Hipocampo/metabolismo , Acontecimientos que Cambian la Vida , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Estrés Psicológico , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Ansiedad , Trastornos de Ansiedad/diagnóstico , Conducta Animal , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Genotipo , Hipocampo/fisiopatología , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Fosforilación , Unión Proteica , Evaluación de Síntomas , Proteínas de Unión a Tacrolimus/genética
12.
Alzheimers Res Ther ; 11(1): 58, 2019 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-31253191

RESUMEN

BACKGROUND: Tau stabilizes microtubules; however, in Alzheimer's disease (AD) and tauopathies, tau becomes hyperphosphorylated, aggregates, and results in neuronal death. Our group recently uncovered a unique interaction between polyamine metabolism and tau fate. Polyamines exert an array of physiological effects that support neuronal function and cognitive processing. Specific stimuli can elicit a polyamine stress response (PSR), resulting in altered central polyamine homeostasis. Evidence suggests that elevations in polyamines following a short-term stressor are beneficial; however, persistent stress and subsequent PSR activation may lead to maladaptive polyamine dysregulation, which is observed in AD, and may contribute to neuropathology and disease progression. METHODS: Male and female mice harboring tau P301L mutation (rTg4510) were examined for a tau-induced central polyamine stress response (tau-PSR). The direct effect of tau-PSR byproducts on tau fibrillization and oligomerization were measured using a thioflavin T assay and a N2a split superfolder GFP-Tau (N2a-ssGT) cell line, respectively. To therapeutically target the tau-PSR, we bilaterally injected caspase 3-cleaved tau truncated at aspartate 421 (AAV9 Tau ΔD421) into the hippocampus and cortex of spermidine/spermine-N1-acetyltransferase (SSAT), a key regulator of the tau-PSR, knock out (SSAT-/-), and wild type littermates, and the effects on tau neuropathology, polyamine dysregulation, and behavior were measured. Lastly, cellular models were employed to further examine how SSAT repression impacted tau biology. RESULTS: Tau induced a unique tau-PSR signature in rTg4510 mice, notably in the accumulation of acetylated spermidine. In vitro, higher-order polyamines prevented tau fibrillization but acetylated spermidine failed to mimic this effect and even promoted fibrillization and oligomerization. AAV9 Tau ΔD421 also elicited a unique tau-PSR in vivo, and targeted disruption of SSAT prevented the accumulation of acetylated polyamines and impacted several tau phospho-epitopes. Interestingly, SSAT knockout mice presented with altered behavior in the rotarod task, the elevated plus maze, and marble burying task, thus highlighting the impact of polyamine homeostasis within the brain. CONCLUSION: These data represent a novel paradigm linking tau pathology and polyamine dysfunction and that targeting specific arms within the polyamine pathway may serve as new targets to mitigate certain components of the tau phenotype.


Asunto(s)
Acetiltransferasas/metabolismo , Poliaminas/metabolismo , Estrés Fisiológico , Tauopatías/enzimología , Acetiltransferasas/genética , Animales , Femenino , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Agregación Patológica de Proteínas/metabolismo , Proteínas tau/metabolismo
13.
eNeuro ; 6(1)2019.
Artículo en Inglés | MEDLINE | ID: mdl-30963102

RESUMEN

Increased expression of the FK506-binding protein 5 (FKBP5) gene has been associated with a number of diseases, but most prominently in connection to psychiatric illnesses. Many of these psychiatric disorders present with dementia and other cognitive deficits, but a direct connection between these issues and alterations in FKBP5 remains unclear. We generated a novel transgenic mouse to selectively overexpress FKBP5, which encodes the FKBP51 protein, in the corticolimbic system, which had no overt effects on gross body weight, motor ability, or general anxiety. Instead, we found that overexpression of FKBP51 impaired long-term depression (LTD) as well as spatial reversal learning and memory, suggesting a role in glutamate receptor regulation. Indeed, FKBP51 altered the association of heat-shock protein 90 (Hsp90) with AMPA receptors, which was accompanied by an accelerated rate of AMPA recycling. In this way, the chaperone system is critical in triage decisions for AMPA receptor trafficking. Imbalance in the chaperone system may manifest in impairments in both inhibitory learning and cognitive function. These findings uncover an unexpected and essential mechanism for learning and memory that is controlled by the psychiatric risk factor FKBP5.


Asunto(s)
Cognición/fisiología , Disfunción Cognitiva/metabolismo , Receptores AMPA/metabolismo , Aprendizaje Espacial/fisiología , Proteínas de Unión a Tacrolimus/biosíntesis , Animales , Disfunción Cognitiva/patología , Femenino , Humanos , Depresión Sináptica a Largo Plazo/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Transporte de Proteínas/fisiología
14.
Int J Biol Macromol ; 127: 136-145, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30639592

RESUMEN

A microsatellite expansion mutation in C9orf72 is the most common genetic cause of Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD). The expansion mutation leads to C9orf72 loss of function, RNA foci formation, and generation of five species of non-AUG RAN translated dipeptide repeat proteins (DPRs), such as poly(GA), poly(GP), poly(GR), poly(PA), and poly(PR). Although one cell can contain more than type of DPRs, information about interplay between different DPR species is limited. Here we show that the combined expression of distinct C9orf72-derived dipeptide repeat species produces cellular outcomes and structural differences that are unique compared to the expression of a single DPR species, suggesting the complex biological interactions that occur when multiple DPR variants are simultaneously expressed. Our data highlights the need for further analysis of how combined expression of different DPRs affects the disease state.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteína C9orf72 , Dipéptidos , Demencia Frontotemporal , Secuencias Repetitivas de Aminoácido , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Línea Celular , Dipéptidos/genética , Dipéptidos/metabolismo , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Humanos
15.
Nat Commun ; 9(1): 4532, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30382094

RESUMEN

The molecular chaperone Hsp90 is critical for the maintenance of cellular homeostasis and represents a promising drug target. Despite increasing knowledge on the structure of Hsp90, the molecular basis of substrate recognition and pro-folding by Hsp90/co-chaperone complexes remains unknown. Here, we report the solution structures of human full-length Hsp90 in complex with the PPIase FKBP51, as well as the 280 kDa Hsp90/FKBP51 complex bound to the Alzheimer's disease-related protein Tau. We reveal that the FKBP51/Hsp90 complex, which synergizes to promote toxic Tau oligomers in vivo, is highly dynamic and stabilizes the extended conformation of the Hsp90 dimer resulting in decreased Hsp90 ATPase activity. Within the ternary Hsp90/FKBP51/Tau complex, Hsp90 serves as a scaffold that traps the PPIase and nucleates multiple conformations of Tau's proline-rich region next to the PPIase catalytic pocket in a phosphorylation-dependent manner. Our study defines a conceptual model for dynamic Hsp90/co-chaperone/client recognition.


Asunto(s)
Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/toxicidad , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/toxicidad , Proteínas tau/química , Proteínas tau/toxicidad , Biocatálisis/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Conformación Proteica , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas tau/metabolismo
16.
ACS Chem Biol ; 13(8): 2288-2299, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29893552

RESUMEN

Genetic and epigenetic alterations in FK506-binding protein 5 ( FKBP5) have been associated with increased risk for psychiatric disorders, including post-traumatic stress disorder (PTSD). Some of these common variants can increase the expression of FKBP5, the gene that encodes FKBP51. Excess FKBP51 promotes hypothalamic-pituitary-adrenal (HPA) axis dysregulation through altered glucocorticoid receptor (GR) signaling. Thus, we hypothesized that GR activity could be restored by perturbing FKBP51. Here, we screened 1280 pharmacologically active compounds and identified three compounds that rescued FKBP51-mediated suppression of GR activity without directly activating GR. One of the three compounds, benztropine mesylate, disrupted the association of FKBP51 with the GR/Hsp90 complex in vitro. Moreover, we show that removal of FKBP51 from this complex by benztropine restored GR localization in ex vivo brain slices and primary neurons from mice. In conclusion, we have identified a novel disruptor of the FKBP51/GR/Hsp90 complex. Targeting this complex may be a viable approach to developing treatments for disorders related to aberrant FKBP51 expression.


Asunto(s)
Benzotropina/farmacología , Depresión/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/metabolismo , Receptores de Glucocorticoides/metabolismo , Trastornos por Estrés Postraumático/tratamiento farmacológico , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Benzotropina/química , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Depresión/metabolismo , Descubrimiento de Drogas , Humanos , Ratones , Terapia Molecular Dirigida , Unión Proteica/efectos de los fármacos , Trastornos por Estrés Postraumático/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores
17.
Nat Struct Mol Biol ; 25(5): 384-393, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29728653

RESUMEN

A network of molecular chaperones is known to bind proteins ('clients') and balance their folding, function and turnover. However, it is often unclear which chaperones are critical for selective recognition of individual clients. It is also not clear why these key chaperones might fail in protein-aggregation diseases. Here, we utilized human microtubule-associated protein tau (MAPT or tau) as a model client to survey interactions between ~30 purified chaperones and ~20 disease-associated tau variants (~600 combinations). From this large-scale analysis, we identified human DnaJA2 as an unexpected, but potent, inhibitor of tau aggregation. DnaJA2 levels were correlated with tau pathology in human brains, supporting the idea that it is an important regulator of tau homeostasis. Of note, we found that some disease-associated tau variants were relatively immune to interactions with chaperones, suggesting a model in which avoiding physical recognition by chaperone networks may contribute to disease.


Asunto(s)
Proteínas del Choque Térmico HSP40/metabolismo , Agregación Patológica de Proteínas/prevención & control , Proteínas tau/metabolismo , Encéfalo/metabolismo , Humanos , Agregado de Proteínas/fisiología , Unión Proteica/fisiología
18.
ACS Chem Biol ; 13(4): 933-941, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29402077

RESUMEN

Gain-of-function mutations within the olfactomedin (OLF) domain of myocilin result in its toxic intracellular accumulation and hasten the onset of open-angle glaucoma. The absence of myocilin does not cause disease; therefore, strategies aimed at eliminating myocilin could lead to a successful glaucoma treatment. The endoplasmic reticulum Hsp90 paralog Grp94 accelerates OLF aggregation. Knockdown or pharmacological inhibition of Grp94 in cells facilitates clearance of mutant myocilin via a non-proteasomal pathway. Here, we expanded our support for targeting Grp94 over cytosolic paralogs Hsp90α and Hsp90ß. We then developed a high-throughput screening assay to identify new chemical matter capable of disrupting the Grp94/OLF interaction. When applied to a blind, focused library of 17 Hsp90 inhibitors, our miniaturized single-read in vitro thioflavin T -based kinetics aggregation assay exclusively identified compounds that target the chaperone N-terminal nucleotide binding site. In follow up studies, one compound (2) decreased the extent of co-aggregation of Grp94 with OLF in a dose-dependent manner in vitro, and enabled clearance of the aggregation-prone full-length myocilin variant I477N in cells without inducing the heat shock response or causing cytotoxicity. Comparison of the co-crystal structure of compound 2 and another non-selective hit in complex with the N-terminal domain of Grp94 reveals a docking mode tailored to Grp94 and explains its selectivity. A new lead compound has been identified, supporting a targeted chemical biology assay approach to develop a protein degradation-based therapy for myocilin-associated glaucoma by selectively inhibiting Grp94.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Glaucoma/tratamiento farmacológico , Glicoproteínas/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Glicoproteínas de Membrana/antagonistas & inhibidores , Cristalografía por Rayos X , Proteínas del Citoesqueleto , Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo , Glicoproteínas/genética , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Simulación del Acoplamiento Molecular
19.
Sci Rep ; 7(1): 17951, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29263415

RESUMEN

The heat shock protein 90 (Hsp90) family of molecular chaperones regulates protein homeostasis, folding, and degradation. The ER-resident Hsp90 isoform, glucose-regulated protein 94 (Grp94), promotes the aggregation of mutant forms of myocilin, a protein associated with primary open-angle glaucoma. While inhibition of Grp94 promotes the degradation of mutant myocilin in vitro, to date no Grp94-selective inhibitors have been investigated in vivo. Here, a Grp94-selective inhibitor facilitated mutant myocilin degradation and rescued phenotypes in a transgenic mouse model of hereditary primary open-angle glaucoma. Ocular toxicities previously associated with pan-Hsp90 inhibitors were not evident with our Grp94-selective inhibitor, 4-Br-BnIm. Our study suggests that selective inhibition of a distinct Hsp90 family member holds translational promise for ocular and other diseases associated with cell stress and protein misfolding.


Asunto(s)
Glaucoma de Ángulo Abierto/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones Endogámicos C57BL , Ratones Transgénicos
20.
Proc Natl Acad Sci U S A ; 114(36): 9707-9712, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28827321

RESUMEN

The microtubule-associated protein tau (MAPT, tau) forms neurotoxic aggregates that promote cognitive deficits in tauopathies, the most common of which is Alzheimer's disease (AD). The 90-kDa heat shock protein (Hsp90) chaperone system affects the accumulation of these toxic tau species, which can be modulated with Hsp90 inhibitors. However, many Hsp90 inhibitors are not blood-brain barrier-permeable, and several present associated toxicities. Here, we find that the cochaperone, activator of Hsp90 ATPase homolog 1 (Aha1), dramatically increased the production of aggregated tau. Treatment with an Aha1 inhibitor, KU-177, dramatically reduced the accumulation of insoluble tau. Aha1 colocalized with tau pathology in human brain tissue, and this association positively correlated with AD progression. Aha1 overexpression in the rTg4510 tau transgenic mouse model promoted insoluble and oligomeric tau accumulation leading to a physiological deficit in cognitive function. Overall, these data demonstrate that Aha1 contributes to tau fibril formation and neurotoxicity through Hsp90. This suggests that therapeutics targeting Aha1 may reduce toxic tau oligomers and slow or prevent neurodegenerative disease progression.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Chaperonas Moleculares/antagonistas & inhibidores , Chaperonas Moleculares/genética , Agregado de Proteínas , Agregación Patológica de Proteínas/etiología , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/prevención & control , Tauopatías/etiología , Tauopatías/metabolismo , Tauopatías/prevención & control , Proteínas tau/química , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA