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













Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
J Biol Chem ; 293(27): 10796-10809, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29764935

RESUMEN

Hsp70 chaperones bind to various protein substrates for folding, trafficking, and degradation. Considerable structural information is available about how prokaryotic Hsp70 (DnaK) binds substrates, but less is known about mammalian Hsp70s, of which there are 13 isoforms encoded in the human genome. Here, we report the interaction between the human Hsp70 isoform heat shock cognate 71-kDa protein (Hsc70 or HSPA8) and peptides derived from the microtubule-associated protein Tau, which is linked to Alzheimer's disease. For structural studies, we used an Hsc70 construct (called BETA) comprising the substrate-binding domain but lacking the lid. Importantly, we found that truncating the lid does not significantly impair Hsc70's chaperone activity or allostery in vitro Using NMR, we show that BETA is partially dynamically disordered in the absence of substrate and that binding of the Tau sequence GKVQIINKKG (with a KD = 500 nm) causes dramatic rigidification of BETA. NOE distance measurements revealed that Tau binds to the canonical substrate-binding cleft, similar to the binding observed with DnaK. To further develop BETA as a tool for studying Hsc70 interactions, we also measured BETA binding in NMR and fluorescent competition assays to peptides derived from huntingtin, insulin, a second Tau-recognition sequence, and a KFERQ-like sequence linked to chaperone-mediated autophagy. We found that the insulin C-peptide binds BETA with high affinity (KD < 100 nm), whereas the others do not (KD > 100 µm). Together, our findings reveal several similarities and differences in how prokaryotic and mammalian Hsp70 isoforms interact with different substrate peptides.


Asunto(s)
Proteínas del Choque Térmico HSC70/metabolismo , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Proteínas tau/metabolismo , Adenosina Trifosfato/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Sitios de Unión , Cristalografía por Rayos X , Proteínas del Choque Térmico HSC70/química , Proteínas del Choque Térmico HSC70/genética , Humanos , Unión Proteica , Conformación Proteica , Proteínas tau/química , Proteínas tau/genética
4.
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
5.
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
6.
PLoS Biol ; 15(6): e2001336, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28654636

RESUMEN

The accumulation of amyloidogenic proteins is a pathological hallmark of neurodegenerative disorders. The aberrant accumulation of the microtubule associating protein tau (MAPT, tau) into toxic oligomers and amyloid deposits is a primary pathology in tauopathies, the most common of which is Alzheimer's disease (AD). Intrinsically disordered proteins, like tau, are enriched with proline residues that regulate both secondary structure and aggregation propensity. The orientation of proline residues is regulated by cis/trans peptidyl-prolyl isomerases (PPIases). Here we show that cyclophilin 40 (CyP40), a PPIase, dissolves tau amyloids in vitro. Additionally, CyP40 ameliorated silver-positive and oligomeric tau species in a mouse model of tau accumulation, preserving neuronal health and cognition. Nuclear magnetic resonance (NMR) revealed that CyP40 interacts with tau at sites rich in proline residues. CyP40 was also able to interact with and disaggregate other aggregating proteins that contain prolines. Moreover, CyP40 lacking PPIase activity prevented its capacity for disaggregation in vitro. Finally, we describe a unique structural property of CyP40 that may permit disaggregation to occur in an energy-independent manner. This study identifies a novel human protein disaggregase and, for the first time, demonstrates its capacity to dissolve intracellular amyloids.


Asunto(s)
Amiloide/metabolismo , Ciclofilinas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Amiloide/genética , Amiloide/ultraestructura , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/fisiopatología , Peptidil-Prolil Isomerasa F , Ciclofilinas/genética , Ciclosporina/farmacología , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Masculino , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Enfermedades Neurodegenerativas/genética , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas , Tauopatías/genética , Tauopatías/metabolismo , alfa-Sinucleína/genética , Proteínas tau/genética
7.
EMBO J ; 35(14): 1537-49, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27261198

RESUMEN

It is now known that proteins associated with neurodegenerative disease can spread throughout the brain in a prionlike manner. However, the mechanisms regulating the trans-synaptic spread propagation, including the neuronal release of these proteins, remain unknown. The interaction of neurodegenerative disease-associated proteins with the molecular chaperone Hsc70 is well known, and we hypothesized that much like disaggregation, refolding, degradation, and even normal function, Hsc70 may dictate the extracellular fate of these proteins. Here, we show that several proteins, including TDP-43, α-synuclein, and the microtubule-associated protein tau, can be driven out of the cell by an Hsc70 co-chaperone, DnaJC5. In fact, DnaJC5 overexpression induced tau release in cells, neurons, and brain tissue, but only when activity of the chaperone Hsc70 was intact and when tau was able to associate with this chaperone. Moreover, release of tau from neurons was reduced in mice lacking the DnaJC5 gene and when the complement of DnaJs in the cell was altered. These results demonstrate that the dynamics of DnaJ/Hsc70 complexes are critically involved in the release of neurodegenerative disease proteins.


Asunto(s)
Proteínas del Choque Térmico HSC70/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas tau/metabolismo , Línea Celular , Proteínas de Unión al ADN/metabolismo , Humanos , alfa-Sinucleína/metabolismo
8.
ACS Chem Biol ; 11(7): 2041-8, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27177119

RESUMEN

Three scaffolds with inhibitory activity against the heat shock protein 70 (Hsp70) family of chaperones have been found to enhance the degradation of the microtubule associated protein tau in cells, neurons, and brain tissue. This is important because tau accumulation is linked to neurodegenerative diseases including Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Here, we expanded upon this study to investigate the anti-tau efficacy of additional scaffolds with Hsp70 inhibitory activity. Five of the nine scaffolds tested lowered tau levels, with the rhodacyanine and phenothiazine scaffolds exhibiting the highest potency as previously described. Because phenothiazines also inhibit tau aggregation in vitro, we suspected that this activity might be a more accurate predictor of tau lowering. Interestingly, the rhodacyanines did inhibit in vitro tau aggregation to a similar degree as phenothiazines, correlating well with tau-lowering efficacy in cells and ex vivo slices. Moreover, other Hsp70 inhibitor scaffolds with weaker tau-lowering activity in cells inhibited tau aggregation in vitro, albeit at lower potencies. When we tested six well-characterized tau aggregation inhibitors, we determined that this mechanism of action was not a better predictor of tau-lowering than Hsp70 inhibition. Instead, we found that compounds possessing both activities were the most effective at promoting tau clearance. Moreover, cytotoxicity and PAINS activity are critical factors that can lead to false-positive lead identification. Strategies designed around these principles will likely yield more efficacious tau-lowering compounds.


Asunto(s)
Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas tau/antagonistas & inhibidores , Células HEK293 , Humanos , Técnicas In Vitro
9.
Biochemistry ; 54(43): 6576-85, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26451841

RESUMEN

Lung infection with Pseudomonas aeruginosa is the leading cause of death among cystic fibrosis patients. To initiate infection, P. aeruginosa assembles a protein nanomachine, the type III secretion system (T3SS), to inject bacterial proteins directly into target host cells. An important regulator of the P. aeruginosa T3SS is the chaperone protein PcrG, which forms a complex with the tip protein, PcrV. In addition to its role as a chaperone to the tip protein, PcrG also regulates protein secretion. PcrG homologues are also important in the T3SS of other pathogens such as Yersinia pestis, the causative agent of bubonic plague. The atomic structure of PcrG or any member of the family of tip protein chaperones is currently unknown. Here, we show by circular dichroism and nuclear magnetic resonance (NMR) spectroscopy that PcrG lacks a tertiary structure. However, it is not completely disordered but contains secondary structures dominated by two long α-helices from residue 16 to 41 and from residue 55 to 76. The helices of PcrG are partially formed, have similar backbone dynamics, and are flexible. NMR titrations show that the entire length of PcrG residues from position 9 to 76 is involved in binding to PcrV. PcrG adds to the growing list of partially folded or unstructured proteins with important roles in type III secretion.


Asunto(s)
Proteínas Bacterianas/química , Pseudomonas aeruginosa/química , Sistemas de Secreción Tipo III/química , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Dicroismo Circular , Humanos , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/fisiología , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Desnaturalización Proteica , Estructura Secundaria de Proteína , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/fisiología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/fisiología
10.
Hum Mol Genet ; 24(14): 3971-81, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25882706

RESUMEN

The pathological accumulation of abnormally hyperphosphorylated and aggregated tau, a neuronal microtubule (MT)-associated protein that functions to maintain MT stability, is implicated in a number of hereditary and sporadic neurodegenerative diseases including frontotemporal dementia and Alzheimer's disease. Targeting tau for the treatment of these diseases is an area of intense interest and toward that end, modulation of cellular molecular chaperones is a potential therapeutic target. In particular, the constitutive Hsp70 isoform, Hsc70, seems highly interconnected with tau, preserving tau protein levels and synergizing with it to assemble MTs. But the relationship between tau and Hsc70, as well as the impact of this interaction in neurons and its therapeutic implications remain unknown. Using a human dominant negative Hsc70 that resembles isoform selective inhibition of this important chaperone, we found for the first time that Hsc70 activity is required to stimulate MT assembly in cells and brain. However, surprisingly, active Hsc70 also requires active tau to regulate MT assembly in vivo, suggesting that tau acts in some ways as a co-chaperone for Hsc70 to coordinate MT assembly. This was despite tau binding to Hsc70 as substrate, as determined biochemically. Moreover, we show that while chronic Hsc70 inhibition damaged MT dynamics, intermittent treatment with a small molecule Hsp70 inhibitor lowered tau in brain tissue without disrupting MT integrity. Thus, in tauopathies, where MT injury would be detrimental to neurons, the unique relationship of tau with the Hsc70 machinery can be exploited to deplete tau levels without damaging MT networks.


Asunto(s)
Proteínas del Choque Térmico HSC70/metabolismo , Microtúbulos/metabolismo , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Proteínas del Choque Térmico HSC70/genética , Humanos , Espectroscopía de Resonancia Magnética , Ratones Noqueados , Neuronas/metabolismo , Oocitos , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Tauopatías/genética , Tauopatías/terapia , Xenopus , Proteínas tau/genética
11.
J Biol Chem ; 290(21): 13115-27, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25864199

RESUMEN

The constitutively expressed heat shock protein 70 kDa (Hsc70) is a major chaperone protein responsible for maintaining proteostasis, yet how its structure translates into functional decisions regarding client fate is still unclear. We previously showed that Hsc70 preserved aberrant Tau, but it remained unknown if selective inhibition of the activity of this Hsp70 isoform could facilitate Tau clearance. Using single point mutations in the nucleotide binding domain, we assessed the effect of several mutations on the functions of human Hsc70. Biochemical characterization revealed that one mutation abolished both Hsc70 ATPase and refolding activities. This variant resembled the ADP-bound conformer at all times yet remained able to interact with cofactors, nucleotides, and substrates appropriately, resembling a dominant negative Hsc70 (DN-Hsc70). We then assessed the effects of this DN-Hsc70 on its client Tau. DN-Hsc70 potently facilitated Tau clearance via the proteasome in cells and brain tissue, in contrast to wild type Hsc70 that stabilized Tau. Thus, DN-Hsc70 mimics the action of small molecule pan Hsp70 inhibitors with regard to Tau metabolism. This shift in Hsc70 function by a single point mutation was the result of a change in the chaperome associated with Hsc70 such that DN-Hsc70 associated more with Hsp90 and DnaJ proteins, whereas wild type Hsc70 was more associated with other Hsp70 isoforms. Thus, isoform-selective targeting of Hsc70 could be a viable therapeutic strategy for tauopathies and possibly lead to new insights in chaperone complex biology.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas del Choque Térmico HSC70/antagonistas & inhibidores , Proteínas del Choque Térmico HSC70/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Proteínas tau/metabolismo , Western Blotting , Células Cultivadas , Citosol/metabolismo , Polarización de Fluorescencia , Técnica del Anticuerpo Fluorescente , Proteínas del Choque Térmico HSC70/genética , Humanos , Espectroscopía de Resonancia Magnética , Mutación/genética , Unión Proteica , Conformación Proteica , Isoformas de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteínas tau/genética
12.
Acta Neuropathol Commun ; 3: 8, 2015 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-25775028

RESUMEN

INTRODUCTION: The blood-brain barrier (BBB) is damaged in tauopathies, including progressive supranuclear palsy (PSP) and Alzheimer's disease (AD), which is thought to contribute to pathogenesis later in the disease course. In AD, BBB dysfunction has been associated with amyloid beta (Aß) pathology, but the role of tau in this process is not well characterized. Since increased BBB permeability is found in tauopathies without Aß pathology, like PSP, we suspected that tau accumulation alone could not only be sufficient, but even more important than Aß for BBB damage. RESULTS: Longitudinal evaluation of brain tissue from the tetracycline-regulatable rTg4510 tau transgenic mouse model showed progressive IgG, T cell and red blood cell infiltration. The Evans blue (EB) dye that is excluded from the brain when the BBB is intact also permeated the brains of rTg4510 mice following peripheral administration, indicative of a bonafide BBB defect, but this was only evident later in life. Thus, despite the marked brain atrophy and inflammation that occurs earlier in this model, BBB integrity is maintained. Interestingly, BBB dysfunction emerged at the same time that perivascular tau emerged around major hippocampal blood vessels. However, when tau expression was suppressed using doxycycline, BBB integrity was preserved, suggesting that the BBB can be stabilized in a tauopathic brain by reducing tau levels. CONCLUSIONS: For the first time, these data demonstrate that tau alone can initiate breakdown of the BBB, but the BBB is remarkably resilient, maintaining its integrity in the face of marked brain atrophy, neuroinflammation and toxic tau accumulation. Moreover, the BBB can recover integrity when tau levels are reduced. Thus, late stage interventions targeting tau may slow the vascular contributions to cognitive impairment and dementia that occur in tauopathies.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Regulación de la Expresión Génica/genética , Tauopatías/patología , Proteínas tau/deficiencia , Factores de Edad , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Doxiciclina/farmacología , Eritrocitos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inmunoglobulina G/metabolismo , Estudios Longitudinales , Ratones , Ratones Transgénicos , Linfocitos T/patología , Proteínas tau/genética
13.
J Neuroinflammation ; 11: 152, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25183004

RESUMEN

BACKGROUND: Abnormal tau hyperphosphorylation and its accumulation into intra-neuronal neurofibrillary tangles are linked to neurodegeneration in Alzheimer's disease and similar tauopathies. One strategy to reduce accumulation is through immunization, but the most immunogenic tau epitopes have so far remained unknown. To fill this gap, we immunized mice with recombinant tau to build a map of the most immunogenic tau epitopes. METHODS: Non-transgenic and rTg4510 tau transgenic mice aged 5 months were immunized with either human wild-type tau (Wt, 4R0N) or P301L tau (4R0N). Each protein was formulated in Quil A adjuvant. Sera and splenocytes of vaccinated mice were collected to assess the humoral and cellular immune responses to tau. We employed a peptide array assay to identify the most effective epitopes. Brain histology was utilized to measure the effects of vaccination on tau pathology and inflammation. RESULTS: Humoral immune responses following immunization demonstrated robust antibody titers (up to 1:80,000 endpoint titers) to each tau species in both mice models. The number of IFN-γ producing T cells and their proliferation were also increased in splenocytes from immunized mice, indicating an increased cellular immune response, and tau levels and neuroinflammation were both reduced. We identified five immunogenic motifs within either the N-terminal (9-15 and 21-27 amino acids), proline rich (168-174 and 220-228 amino acids), or the C-terminal regions (427-438 amino acids) of the wild-type and P301L tau protein sequence. CONCLUSIONS: Our study identifies five previously unknown immunogenic motifs of wild-type and mutated (P301L) tau protein. Immunization with both proteins resulted in reduced tau pathology and neuroinflammation in a tau transgenic model, supporting the efficacy of tau immunotherapy in tauopathy.


Asunto(s)
Mapeo Epitopo , Epítopos/inmunología , Tauopatías/inmunología , Tauopatías/terapia , Vacunación/métodos , Proteínas tau/inmunología , Adyuvantes Inmunológicos/uso terapéutico , Animales , Anticuerpos/sangre , Proliferación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/etiología , Encefalitis/inmunología , Encefalitis/terapia , Ensayo de Inmunoadsorción Enzimática , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Saponinas de Quillaja , Saponinas/uso terapéutico , Linfocitos T/efectos de los fármacos , Tauopatías/complicaciones , Tauopatías/patología , Proteínas tau/genética
14.
PLoS One ; 9(9): e107241, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25191701

RESUMEN

Single nucleotide polymorphisms (SNPs) in the FK506 binding protein 5 (FKBP5) gene combine with traumatic events to increase risk for post-traumatic stress and major depressive disorders (PTSD and MDD). These SNPs increase FKBP51 protein expression through a mechanism involving demethylation of the gene and altered glucocorticoid signaling. Aged animals also display elevated FKBP51 levels, which contribute to impaired resiliency to depressive-like behaviors through impaired glucocorticoid signaling, a phenotype that is abrogated in FKBP5-/- mice. But the age of onset and progressive stability of these phenotypes remain unknown. Moreover, it is unclear how FKBP5 deletion affects other glucocorticoid-dependent processes or if age-associated increases in FKBP51 expression are mediated through a similar epigenetic process caused by SNPs in the FKBP5 gene. Here, we show that FKBP51-mediated impairment in stress resiliency and glucocorticoid signaling occurs by 10 months of age and this increased over their lifespan. Surprisingly, despite these progressive changes in glucocorticoid responsiveness, FKBP5-/- mice displayed normal longevity, glucose tolerance, blood composition and cytokine profiles across lifespan, phenotypes normally associated with glucocorticoid signaling. We also found that methylation of Fkbp5 decreased with age in mice, a process that likely explains the age-associated increases in FKBP51 levels. Thus, epigenetic upregulation of FKBP51 with age can selectively impair psychological stress-resiliency, but does not affect other glucocorticoid-mediated physiological processes. This makes FKBP51 a unique and attractive therapeutic target to treat PTSD and MDD. In addition, aged wild-type mice may be a useful model for investigating the mechanisms of FKBP5 SNPs associated with these disorders.


Asunto(s)
Envejecimiento/genética , Epigénesis Genética/fisiología , Resiliencia Psicológica , Estrés Psicológico/genética , Proteínas de Unión a Tacrolimus/genética , Envejecimiento/sangre , Animales , Metilación de ADN , Trastorno Depresivo Mayor/genética , Hidrocortisona/sangre , Longevidad/genética , Ratones , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Trastornos por Estrés Postraumático/genética , Estrés Psicológico/sangre , Proteínas de Unión a Tacrolimus/metabolismo , Regulación hacia Arriba/genética
15.
Cell ; 156(5): 963-74, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581495

RESUMEN

Protein folding in the cell relies on the orchestrated action of conserved families of molecular chaperones, the Hsp70 and Hsp90 systems. Hsp70 acts early and Hsp90 late in the folding path, yet the molecular basis of this timing is enigmatic, mainly because the substrate specificity of Hsp90 is poorly understood. Here, we obtained a structural model of Hsp90 in complex with its natural disease-associated substrate, the intrinsically disordered Tau protein. Hsp90 binds to a broad region in Tau that includes the aggregation-prone repeats. Complementarily, a 106-Å-long substrate-binding interface in Hsp90 enables many low-affinity contacts. This allows recognition of scattered hydrophobic residues in late folding intermediates that remain after early burial of the Hsp70 sites. Our model resolves the paradox of how Hsp90 specifically selects for late folding intermediates but also for some intrinsically disordered proteins-through the eyes of Hsp90 they look the same.


Asunto(s)
Proteínas tau/química , Enfermedad de Alzheimer/tratamiento farmacológico , Secuencia de Aminoácidos , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Pliegue de Proteína , Dispersión del Ángulo Pequeño , Difracción de Rayos X , Proteínas tau/metabolismo
16.
J Clin Invest ; 123(10): 4158-69, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23999428

RESUMEN

Aggregation of tau protein in the brain is associated with a class of neurodegenerative diseases known as tauopathies. FK506 binding protein 51 kDa (FKBP51, encoded by FKBP5) forms a mature chaperone complex with Hsp90 that prevents tau degradation. In this study, we have shown that tau levels are reduced throughout the brains of Fkbp5-/- mice. Recombinant FKBP51 and Hsp90 synergized to block tau clearance through the proteasome, resulting in tau oligomerization. Overexpression of FKBP51 in a tau transgenic mouse model revealed that FKBP51 preserved the species of tau that have been linked to Alzheimer's disease (AD) pathogenesis, blocked amyloid formation, and decreased tangle load in the brain. Alterations in tau turnover and aggregate structure corresponded with enhanced neurotoxicity in mice. In human brains, FKBP51 levels increased relative to age and AD, corresponding with demethylation of the regulatory regions in the FKBP5 gene. We also found that higher FKBP51 levels were associated with AD progression. Our data support a model in which age-associated increases in FKBP51 levels and its interaction with Hsp90 promote neurotoxic tau accumulation. Strategies aimed at attenuating FKBP51 levels or its interaction with Hsp90 have the potential to be therapeutically relevant for AD and other tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas tau/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Animales , Región CA3 Hipocampal/metabolismo , Estudios de Casos y Controles , Metilación de ADN , Femenino , Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Complejo de la Endopetidasa Proteasomal/metabolismo , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Proteolisis , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Adulto Joven , Proteínas tau/química
17.
J Neurosci ; 33(22): 9498-507, 2013 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-23719816

RESUMEN

In Alzheimer's disease (AD), the mechanisms of neuronal loss remain largely unknown. Although tau pathology is closely correlated with neuronal loss, how its accumulation may lead to activation of neurotoxic pathways is unclear. Here we show that tau increased the levels of ubiquitinated proteins in the brain and triggered activation of the unfolded protein response (UPR). This suggested that tau interferes with protein quality control in the endoplasmic reticulum (ER). Consistent with this, ubiquitin was found to associate with the ER in human AD brains and tau transgenic (rTg4510) mouse brains, but this was not always colocalized with tau. The increased levels of ubiquitinated protein were accompanied by increased levels of phosphorylated protein kinase R-like ER kinase (pPERK), a marker that indicates UPR activation. Depleting soluble tau levels in cells and brain could reverse UPR activation. Tau accumulation facilitated its deleterious interaction with ER membrane and associated proteins that are essential for ER-associated degradation (ERAD), including valosin-containing protein (VCP) and Hrd1. Based on this, the effects of tau accumulation on ERAD efficiency were evaluated using the CD3δ reporter, an ERAD substrate. Indeed, CD3δ accumulated in both in vitro and in vivo models of tau overexpression and AD brains. These data suggest that soluble tau impairs ERAD and the result is activation of the UPR. The reversibility of this process, however, suggests that tau-based therapeutics could significantly delay this type of cell death and therefore disease progression.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/fisiología , Respuesta de Proteína Desplegada/fisiología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Western Blotting , Encéfalo/patología , Encéfalo/ultraestructura , Química Encefálica , Complejo CD3/metabolismo , Células Cultivadas , Interpretación Estadística de Datos , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Microsomas/metabolismo , Ubiquitina/metabolismo , eIF-2 Quinasa/metabolismo
18.
FASEB J ; 27(4): 1450-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23271055

RESUMEN

Dysfunctional tau accumulation is a major contributing factor in tauopathies, and the heat-shock protein 70 (Hsp70) seems to play an important role in this accumulation. Several reports suggest that Hsp70 proteins can cause tau degradation to be accelerated or slowed, but how these opposing activities are controlled is unclear. Here we demonstrate that highly homologous variants in the Hsp70 family can have opposing effects on tau clearance kinetics. When overexpressed in a tetracycline (Tet)-based protein chase model, constitutive heat shock cognate 70 (Hsc70) and inducible Hsp72 slowed or accelerated tau clearance, respectively. Tau synergized with Hsc70, but not Hsp72, to promote microtubule assembly at nearly twice the rate of either Hsp70 homologue in reconstituted, ATP-regenerating Xenopus extracts supplemented with rhodamine-labeled tubulin and human recombinant Hsp72 and Hsc70. Nuclear magnetic resonance spectroscopy with human recombinant protein revealed that Hsp72 had greater affinity for tau than Hsc70 (I/I0 ratio difference of 0.3), but Hsc70 was 30 times more abundant than Hsp72 in human and mouse brain tissue. This indicates that the predominant Hsp70 variant in the brain is Hsc70, suggesting that the brain environment primarily supports slower tau clearance. Despite its capacity to clear tau, Hsp72 was not induced in the Alzheimer's disease brain, suggesting a mechanism for age-associated onset of the disease. Through the use of chimeras that blended the domains of Hsp72 and Hsc70, we determined that the reason for these differences between Hsc70 and Hsp72 with regard to tau clearance kinetics lies within their C-terminal domains, which are essential for their interactions with substrates and cochaperones. Hsp72 but not Hsc70 in the presence of tau was able to recruit the cochaperone ubiquitin ligase CHIP, which is known to facilitate the ubiquitination of tau, describing a possible mechanism of how the C-termini of these homologous Hsp70 variants can differentially regulate tau triage. Thus, efforts to promote Hsp72 expression and inhibit Hsc70 could be therapeutically relevant for tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas del Choque Térmico HSP72/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Animales , Proteínas del Choque Térmico HSC70/genética , Proteínas del Choque Térmico HSP72/genética , Humanos , Ratones , Unión Proteica/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas tau/genética
19.
Protein Sci ; 20(1): 75-86, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21031487

RESUMEN

The type III secretion system (T3SS) is a protein injection nanomachinery required for virulence by many human pathogenic bacteria including Salmonella and Shigella. An essential component of the T3SS is the tip protein and the Salmonella SipD and the Shigella IpaD tip proteins interact with bile salts, which serve as environmental sensors for these enteric pathogens. SipD and IpaD have long central coiled coils and their N-terminal regions form α-helical hairpins and a short helix α3 that pack against the coiled coil. Using AutoDock, others have predicted that the bile salt deoxycholate binds IpaD in a cleft formed by the α-helical hairpin and its long central coiled coil. NMR chemical shift mapping, however, indicated that the SipD residues most affected by bile salts are located in a disordered region near helix α3. Thus, how bile salts interact with SipD and IpaD is unclear. Here, we report the crystal structures of SipD in complex with the bile salts deoxycholate and chenodeoxycholate. Bile salts bind SipD in a region different from what was predicted for IpaD. In SipD, bile salts bind part of helix α3 and the C-terminus of the long central coiled coil, towards the C-terminus of the protein. We discuss the biological implication of the differences in how bile salts interact with SipD and IpaD.


Asunto(s)
Antígenos Bacterianos/química , Proteínas Bacterianas/química , Sistemas de Secreción Bacterianos , Ácido Quenodesoxicólico/química , Ácido Desoxicólico/química , Proteínas de la Membrana/química , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Células Cultivadas , Ácido Quenodesoxicólico/metabolismo , Dicroismo Circular , Cristalografía por Rayos X , Ácido Desoxicólico/metabolismo , Células Epiteliales/microbiología , Humanos , Espectroscopía de Resonancia Magnética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad
20.
Biochemistry ; 49(19): 4220-6, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20397637

RESUMEN

Salmonella and Shigella bacteria require the type III secretion system (T3SS) to inject virulence proteins into their hosts and initiate infections. The tip proteins SipD and IpaD are critical components of the Salmonella and Shigella T3SS, respectively. Recently, SipD and IpaD have been shown to interact with bile salts, which are enriched in the intestines, and are hypothesized to act as environmental sensors for these enteric pathogens. Bile salts activate the Shigella T3SS but repress the Salmonella T3SS, and the mechanism of this differing response to bile salts is poorly understood. Further, how SipD binds to bile salts is currently unknown. Computer modeling predicted that IpaD binds the bile salt deoxycholate in a cleft formed by the N-terminal domain and the long central coiled coil of IpaD. Here, we used NMR methods to determine which SipD residues are affected by the interaction with the bile salts deoxycholate, chenodeoxycholate, and taurodeoxcholate. The bile salts perturbed nearly the same set of SipD residues; however, the largest chemical shift perturbations occurred away from what was predicted for the bile salt binding site in IpaD. Our NMR results indicate that that bile salt interaction of SipD will be different from what was predicted for IpaD, suggesting a possible mechanism for the differing response of Salmonella and Shigella to bile salts.


Asunto(s)
Antígenos Bacterianos/química , Proteínas Bacterianas/química , Ácidos y Sales Biliares/metabolismo , Proteínas de la Membrana/química , Salmonella/metabolismo , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA