Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
EMBO Rep ; 21(3): e48328, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31930681

RESUMEN

Overexpressing Tau counteracts apoptosis and increases dephosphorylated ß-catenin levels, but the underlying mechanisms are elusive. Here, we show that Tau can directly and robustly acetylate ß-catenin at K49 in a concentration-, time-, and pH-dependent manner. ß-catenin K49 acetylation inhibits its phosphorylation and its ubiquitination-associated proteolysis, thus increasing ß-catenin protein levels. K49 acetylation further promotes nuclear translocation and the transcriptional activity of ß-catenin, and increases the expression of survival-promoting genes (bcl2 and survivin), counteracting apoptosis. Mutation of Tau's acetyltransferase domain or co-expressing non-acetylatable ß-catenin-K49R prevents increased ß-catenin signaling and abolishes the anti-apoptotic function of Tau. Our data reveal that Tau preserves ß-catenin by acetylating K49, and upregulated ß-catenin/survival signaling in turn mediates the anti-apoptotic effect of Tau.


Asunto(s)
Transducción de Señal , beta Catenina , Proteínas tau , Acetilación , Apoptosis/genética , Supervivencia Celular/genética , Humanos , Fosforilación , beta Catenina/genética , beta Catenina/metabolismo
2.
Acta Biochim Biophys Sin (Shanghai) ; 46(2): 136-40, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24374663

RESUMEN

Transcription factor activator protein 2α (AP-2α) is a negative regulator of adipogenesis by repressing the transcription of CCAAT/enhancer binding protein (C/EBPα) gene. During adipogenesis, AP-2α is degraded, leading to transcriptional up-regulation of C/EBPα. However, the mechanism for AP-2α degradation is not clear. Here, using immunoprecipitation assay and mass spectrometry, we identified ring finger protein 20 (RNF20) as an AP-2α-interacting protein in 3T3-L1 preadipocytes. RNF20 has been proved to be an E3 ubiquitin ligase for both histone H2B and tumor suppressor ErbB3-binding protein 1 (Ebp1). In this study, we demonstrated that RNF20 co-localized and interacted with AP-2α, and promoted its polyubiquitination and proteasome-dependent degradation. Over-expression of RNF20 inhibited the activity of AP-2α and rescued the C/EBPα expression which was inhibited by AP-2α. These results suggested that RNF20 may play roles in adipocyte differentiation by stimulating ubiquitin-proteasome-dependent degradation of AP-2α.


Asunto(s)
Factor de Transcripción AP-2/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Adipogénesis/genética , Animales , Proteína alfa Potenciadora de Unión a CCAAT/biosíntesis , Diferenciación Celular , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación
3.
Mil Med Res ; 11(1): 16, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38462603

RESUMEN

BACKGROUND: Episodic memory loss is a prominent clinical manifestation of Alzheimer's disease (AD), which is closely related to tau pathology and hippocampal impairment. Due to the heterogeneity of brain neurons, the specific roles of different brain neurons in terms of their sensitivity to tau accumulation and their contribution to AD-like social memory loss remain unclear. Therefore, further investigation is necessary. METHODS: We investigated the effects of AD-like tau pathology by Tandem mass tag proteomic and phosphoproteomic analysis, social behavioural tests, hippocampal electrophysiology, immunofluorescence staining and in vivo optical fibre recording of GCaMP6f and iGABASnFR. Additionally, we utilized optogenetics and administered ursolic acid (UA) via oral gavage to examine the effects of these agents on social memory in mice. RESULTS: The results of proteomic and phosphoproteomic analyses revealed the characteristics of ventral hippocampal CA1 (vCA1) under both physiological conditions and AD-like tau pathology. As tau progressively accumulated, vCA1, especially its excitatory and parvalbumin (PV) neurons, were fully filled with mislocated and phosphorylated tau (p-Tau). This finding was not observed for dorsal hippocampal CA1 (dCA1). The overexpression of human tau (hTau) in excitatory and PV neurons mimicked AD-like tau accumulation, significantly inhibited neuronal excitability and suppressed distinct discrimination-associated firings of these neurons within vCA1. Photoactivating excitatory and PV neurons in vCA1 at specific rhythms and time windows efficiently ameliorated tau-impaired social memory. Notably, 1 month of UA administration efficiently decreased tau accumulation via autophagy in a transcription factor EB (TFEB)-dependent manner and restored the vCA1 microcircuit to ameliorate tau-impaired social memory. CONCLUSION: This study elucidated distinct protein and phosphoprotein networks between dCA1 and vCA1 and highlighted the susceptibility of the vCA1 microcircuit to AD-like tau accumulation. Notably, our novel findings regarding the efficacy of UA in reducing tau load and targeting the vCA1 microcircuit may provide a promising strategy for treating AD in the future.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Masculino , Ratones , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Ratones Transgénicos , Proteómica , Hipocampo/metabolismo , Hipocampo/patología , Trastornos de la Memoria/metabolismo
4.
J Alzheimers Dis ; 93(1): 169-178, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36970911

RESUMEN

BACKGROUND: Olfactory dysfunction appears prior to cognitive decline, and thus it has been suggested to be an early predictor of Alzheimer's disease. However, it is currently not known whether and how olfactory threshold test could serve as a quick screening tool for cognitive impairment. OBJECTIVE: To define olfactory threshold test for screening cognitive impairment in two independent cohorts. METHODS: The participants are comprised of two cohorts in China, 1,139 inpatients with type 2 diabetes mellitus (T2DM, Discovery cohort) and 1,236 community-dwelling elderly (Validation cohort). Olfactory and cognitive functions were evaluated by Connecticut Chemosensory Clinical Research Center test and Mini-Mental State Examination (MMSE), respectively. Regression analyses and receiver operating characteristic (ROC) analyses were carried out to determine the relation and discriminative performance of the olfactory threshold score (OTS) regarding identification of cognition impairment. RESULTS: Regression analysis showed that olfactory deficit (reducing OTS) was correlated with cognitive impairment (reducing MMSE score) in two cohorts. ROC analysis revealed that the OTS could distinguish cognitive impairment from cognitively normal individuals, with mean area under the curve values of 0.71 (0.67, 0.74) and 0.63 (0.60, 0.66), respectively, but it failed to discriminate dementia from mild cognitive impairment. The cut-off point of 3 showed the highest validity for the screening, with the diagnostic accuracy of 73.3% and 69.5%. CONCLUSION: Reducing OTS is associated with cognitive impairment in T2DM patients and the community-dwelling elderly. Therefore, olfactory threshold test may be used as a readily accessible screening tool for cognitive impairment.


Asunto(s)
Enfermedad de Alzheimer , Trastornos del Conocimiento , Disfunción Cognitiva , Diabetes Mellitus Tipo 2 , Humanos , Anciano , Diabetes Mellitus Tipo 2/complicaciones , Pruebas Neuropsicológicas , Disfunción Cognitiva/psicología , Enfermedad de Alzheimer/psicología , Trastornos del Conocimiento/diagnóstico , Curva ROC , Tamizaje Masivo
5.
Mol Neurodegener ; 18(1): 23, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37060096

RESUMEN

BACKGROUND: Abnormal tau accumulation and cholinergic degeneration are hallmark pathologies in the brains of patients with Alzheimer's disease (AD). However, the sensitivity of cholinergic neurons to AD-like tau accumulation and strategies to ameliorate tau-disrupted spatial memory in terms of neural circuits still remain elusive. METHODS: To investigate the effect and mechanism of the cholinergic circuit in Alzheimer's disease-related hippocampal memory, overexpression of human wild-type Tau (hTau) in medial septum (MS)-hippocampus (HP) cholinergic was achieved by specifically injecting pAAV-EF1α-DIO-hTau-eGFP virus into the MS of ChAT-Cre mice. Immunostaining, behavioral analysis and optogenetic activation experiments were used to detect the effect of hTau accumulation on cholinergic neurons and the MS-CA1 cholinergic circuit. Patch-clamp recordings and in vivo local field potential recordings were used to analyze the influence of hTau on the electrical signals of cholinergic neurons and the activity of cholinergic neural circuit networks. Optogenetic activation combined with cholinergic receptor blocker was used to detect the role of cholinergic receptors in spatial memory. RESULTS: In the present study, we found that cholinergic neurons with an asymmetric discharge characteristic in the MS-hippocampal CA1 pathway are vulnerable to tau accumulation. In addition to an inhibitory effect on neuronal excitability, theta synchronization between the MS and CA1 subsets was significantly disrupted during memory consolidation after overexpressing hTau in the MS. Photoactivating MS-CA1 cholinergic inputs within a critical 3 h time window during memory consolidation efficiently improved tau-induced spatial memory deficits in a theta rhythm-dependent manner. CONCLUSIONS: Our study not only reveals the vulnerability of a novel MS-CA1 cholinergic circuit to AD-like tau accumulation but also provides a rhythm- and time window-dependent strategy to target the MS-CA1 cholinergic circuit, thereby rescuing tau-induced spatial cognitive functions.


Asunto(s)
Enfermedad de Alzheimer , Consolidación de la Memoria , Animales , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Colinérgicos/metabolismo , Colinérgicos/farmacología , Neuronas Colinérgicas , Hipocampo/metabolismo , Trastornos de la Memoria/metabolismo , Proteínas tau/metabolismo
6.
EBioMedicine ; 78: 103970, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35339896

RESUMEN

BACKGROUND: Glycogen synthase kinase-3ß (GSK-3ß) is one of the most effective kinases in promoting tau hyperphosphorylation and accumulation in Alzheimer's disease (AD). However, it is not clear how GSK-3ß activity is regulated during AD progression. METHODS: We firstly used mass spectrometry to identify the acetylation site of GSK-3ß, and then established the cell and animal models of GSK-3ß acetylation. Next, we conducted molecular, cell biological and behavioral tests. Finally, we designed a peptide to test whether blocking tau-mediated GSK-3ß acetylation could be beneficial to AD. FINDINGS: We found that GSK-3ß protein levels increased in the brains of AD patients and the transgenic mice. Overexpressing tau increased GSK-3ß protein level with increased acetylation and decreased ubiquitination-related proteolysis. Tau could directly acetylate GSK-3ß at K15 both in vitro and in vivo. K15-acetylation inhibited ubiquitination-associated proteolysis of GSK-3ß and changed its activity-dependent phosphorylation, leading to over-activation of the kinase. GSK-3ß activation by K15-acetylation in turn exacerbated the AD-like pathologies. Importantly, competitively inhibiting GSK-3ß K15-acetylation by a novel-designed peptide remarkably improved cognitive impairment and the AD-like pathologies in 3xTg-AD mice. INTERPRETATION: Tau can directly acetylate GSK-3ß at K15 which reveals a vicious cycle between tau hyperphosphorylation and GSK-3ß activation. FUNDING: This study was supported in parts by grants from Science and Technology Committee of China (2016YFC1305800), Hubei Province (2018ACA142), Natural Science Foundation of China (91949205, 82001134, 31730035, 81721005), Guangdong Provincial Key S&T Program (018B030336001).


Asunto(s)
Enfermedad de Alzheimer , Proteínas tau , Acetilación , Enfermedad de Alzheimer/metabolismo , Animales , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Ratones , Fosforilación , Proteínas tau/metabolismo
7.
Aging Cell ; 21(5): e13600, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35355405

RESUMEN

Abnormal tau accumulation and spatial memory loss constitute characteristic pathology and symptoms of Alzheimer disease (AD). Yet, the intrinsic connections and the mechanism between them are not fully understood. In the current study, we observed a prominent accumulation of the AD-like hyperphosphorylated and truncated tau (hTau N368) proteins in hippocampal dentate gyrus (DG) mossy cells of 3xTg-AD mice. Further investigation demonstrated that the ventral DG (vDG) mossy cell-specific overexpressing hTau for 3 months induced spatial cognitive deficits, while expressing hTau N368 for only 1 month caused remarkable spatial cognitive impairment with more prominent tau pathologies. By in vivo electrophysiological and optic fiber recording, we observed that the vDG mossy cell-specific overexpression of hTau N368 disrupted theta oscillations with local neural network inactivation in the dorsal DG subset, suggesting impairment of the ventral to dorsal neural circuit. The mossy cell-specific transcriptomic data revealed that multiple AD-associated signaling pathways were disrupted by hTau N368, including reduction of synapse-associated proteins, inhibition of AKT and activation of glycogen synthase kinase-3ß. Importantly, chemogenetic activating mossy cells efficiently attenuated the hTau N368-induced spatial cognitive deficits. Together, our findings indicate that the mossy cell pathological tau accumulation could induce the AD-like spatial memory deficit by inhibiting the local neural network activity, which not only reveals new pathogenesis underlying the mossy cell-related spatial memory loss but also provides a mouse model of Mossy cell-specific hTau accumulation for drug development in AD and the related tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/patología , Animales , Cognición , Disfunción Cognitiva/genética , Modelos Animales de Enfermedad , Trastornos de la Memoria/metabolismo , Ratones , Ratones Transgénicos , Fibras Musgosas del Hipocampo/metabolismo , Fibras Musgosas del Hipocampo/patología , Proteínas tau/metabolismo
8.
Clin Transl Med ; 12(8): e1003, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35917404

RESUMEN

BACKGROUND: Human Tau (hTau) accumulation and synapse loss are two pathological hallmarks of tauopathies. However, whether and how hTau exerts toxic effects on synapses remain elusive. METHODS: Mutated hTau (P301S) was overexpressed in the N2a cell line, primary hippocampal neurons and hippocampal CA3. Western blotting and quantitative polymerase chain reaction were applied to examine the protein and mRNA levels of synaptic proteins. The protein interaction was tested by co-immunoprecipitation and proximity ligation assays. Memory and emotion status were evaluated by a series of behavioural tests. The transcriptional activity of nuclear factor-erythroid 2-related factor 2 (NRF2) was detected by dual luciferase reporter assay. Electrophoresis mobility shift assay and chromosome immunoprecipitation were conducted to examine the combination of NRF2 to specific anti-oxidative response element (ARE) sequences. Neuronal morphology was analysed after Golgi staining. RESULTS: Overexpressing P301S decreased the protein levels of post-synaptic density protein 93 (PSD93), PSD95 and synapsin 1 (SYN1). Simultaneously, NRF2 was decreased, whereas Kelch-like ECH-associated protein 1 (KEAP1) was elevated. Further, we found that NRF2 could bind to the specific AREs of DLG2, DLG4 and SYN1 genes, which encode PSD93, PSD95 and SYN1, respectively, to promote their expression. Overexpressing NRF2 ameliorated P301S-reduced synaptic proteins and synapse. By means of acetylation at K312, P301S increased the protein level of KEAP1 via inhibiting KEAP1 degradation from ubiquitin-proteasome pathway, thereby decreasing NRF2 and reducing synapse. Blocking the P301S-KEAP1 interaction at K312 rescued the P301S-suppressed expression of synaptic proteins and memory deficits with anxiety efficiently. CONCLUSIONS: P301S-hTau could acetylate KEAP1 to trigger synaptic toxicity via inhibiting the NRF2/ARE pathway. These findings provide a novel and potential target for the therapeutic intervention of tauopathies.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Tauopatías , Hidrolasas de Éster Carboxílico/metabolismo , Genes Reguladores , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Elementos de Respuesta , Tauopatías/genética
9.
Mil Med Res ; 9(1): 38, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35799293

RESUMEN

BACKGROUND: Autophagy dysfunction plays a crucial role in tau accumulation and neurodegeneration in Alzheimer's disease (AD). This study aimed to investigate whether and how the accumulating tau may in turn affect autophagy. METHODS: The primary hippocampal neurons, N2a and HEK293T cells with tau overexpression were respectively starved and treated with vinblastine to study the effects of tau on the initiating steps of autophagy, which was analysed by Student's two-tailed t-test. The rapamycin and concanamycin A were employed to inhibit the mammalian target of rapamycin kinase complex 1 (mTORC1) activity and the vacuolar H+-ATPase (v-ATPase) activity, respectively, which were analysed by One-way ANOVA with post hoc tests. The Western blotting, co-immunoprecipitation and immunofluorescence staining were conducted to gain insight into the mechanisms underlying the tau effects of mTORC1 signaling alterations, as analysed by Student's two-tailed t-test or One-way ANOVA with post hoc tests. The autophagosome formation was detected by immunofluorescence staining and transmission electron microscopy. The amino acids (AA) levels were detected by high performance liquid chromatography (HPLC). RESULTS: We observed that overexpressing human full-length wild-type tau to mimic AD-like tau accumulation induced autophagy deficits. Further studies revealed that the increased tau could bind to the prion-related domain of T cell intracellular antigen 1 (PRD-TIA1) and this association significantly increased the intercellular level of amino acids (Leucine, P = 0.0038; Glutamic acid, P = 0.0348; Alanine, P = 0.0037; Glycine, P = 0.0104), with concordant upregulation of mTORC1 activity [phosphorylated eukaryotic translation initiation factor 4E-binding protein 1 (p-4EBP1), P < 0.0001; phosphorylated 70 kDa ribosomal protein S6 kinase 1 (p-p70S6K1), P = 0.0001, phosphorylated unc-51-like autophagy-activating kinase 1 (p-ULK1), P = 0.0015] and inhibition of autophagosome formation [microtubule-associated protein light chain 3 II (LC3 II), P = 0.0073; LC3 puncta, P < 0.0001]. As expected, this tau-induced deficit of autophagosome formation in turn aggravated tau accumulation. Importantly, we also found that blocking TIA1 and tau interaction by overexpressing PRD-TIA1, downregulating the endogenous TIA1 expression by shRNA, or downregulating tau protein level by a small proteolysis targeting chimera (PROTAC) could remarkably attenuate tau-induced autophagy impairment. CONCLUSIONS: Our findings reveal that AD-like tau accumulation inhibits autophagosome formation and induces autophagy deficits by activating the TIA1/amino acid/mTORC1 pathway, and thus this work reveals new insight into tau-associated neurodegeneration and provides evidence supporting the use of new therapeutic targets for AD treatment and that of related tauopathies.


Asunto(s)
Autofagosomas , Diana Mecanicista del Complejo 1 de la Rapamicina , Antígeno Intracelular 1 de las Células T , Proteínas tau , Aminoácidos/metabolismo , Autofagosomas/metabolismo , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Antígeno Intracelular 1 de las Células T/metabolismo , Proteínas tau/metabolismo , Proteínas tau/farmacología
10.
J Alzheimers Dis ; 81(4): 1403-1418, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33935099

RESUMEN

BACKGROUND: Recent studies show that an increased T217-phosphorylation of tau in plasma could diagnose AD at an early stage with high accuracy and high specificity, while the potential toxic role of tau T217-phosphorylation is not known. OBJECTIVE: To study the potential toxic role of tau T217-phosphorylation. METHODS: We performed stereotactic brain injection, behavioral testing, immunohistochemistry and immunofluorescence, western blotting, Golgi staining, in vitro recombinant tau polymerization, and other measurements. RESULTS: We first constructed tau T217-wild-type (T217), T217-phospho-mimic (T217E), and T217-non-phospho-mimic (T217A) plasmids or their virus vectors on the basis of wild-type tau. We found that expressing tau-T217E induced a significantly increased tau phosphorylation at multiple AD-associated sites with inhibited proteolysis and increased cleavage/fibrillization of tau, while expressing tau-T217A abolished the above changes of tau both in vitro and in vivo. By mutating T217E on tau-P301L, a dominant mutation identified in patients with frontotemporal dementia, we did not observe significant exacerbation of tau-P301L phosphorylation and cognitive impairment although the increased tau cleavage and propagation were shown. CONCLUSION: T217-phosphorylation exacerbates wild-type tau hyperphosphorylation with aggravated tau cleavage/fibrillization and cognitive impairments, while overexpressing T217E on the basis P301L does not exacerbate tau phosphorylation or the P301L-induced cognitive deficits, although it aggravates tau cleavage and propagation.


Asunto(s)
Cognición/fisiología , Disfunción Cognitiva/metabolismo , Hipocampo/metabolismo , Aprendizaje por Laberinto/fisiología , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Disfunción Cognitiva/patología , Hipocampo/patología , Humanos , Ratones , Fosforilación , Tauopatías/patología
11.
Theranostics ; 11(11): 5279-5295, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859747

RESUMEN

Intracellular accumulation of tau is a hallmark pathology in Alzheimer disease (AD) and the related tauopathies, thus targeting tau could be promising for drug development. Proteolysis Targeting Chimera (PROTAC) is a novel drug discovery strategy for selective protein degradation from within cells. Methods: A novel small-molecule PROTAC, named as C004019 with a molecular mass of 1,035.29 dalton, was designed to simultaneously recruite tau and E3-ligase (Vhl) and thus to selectively enhance ubiquitination and proteolysis of tau proteins. Western blotting, immunofluoresence and immunohistochemical staining were employed to verify the effects of C004019 in cell models (HEK293 and SH-SY5Y) and mouse models (hTau-transgenic and 3xTg-AD), respectively. The cognitive capacity of the mice was assessed by a suite of behavior experiments. Electrophysiology and Golgi staining were used to evaluate the synaptic plasticity. Results: C004019 induced a robust tau clearance via promoting its ubiquitination-proteasome-dependent proteolysis in HEK293 cells with stable or transient overexpression of human tau (hTau), and in SH-SY5Y that constitutively overexpress hTau. Furthermore, intracerebral ventricular infusion of C004019 induced a robust tau clearance in vivo. Most importantly, both single-dose and multiple-doses (once per 6 days for a total 5 times) subcutaneous administration of C004019 remarkably decreased tau levels in the brains of wild-type, hTau-transgenic and 3xTg-AD mice with improvement of synaptic and cognitive functions. Conclusions: The PROTAC (C004019) created in the current study can selectively and efficiently promote tau clearance both in vitro and in vivo, which provides a promising drug candidate for AD and the related tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Cognición , Bibliotecas de Moléculas Pequeñas , Proteínas tau , Animales , Humanos , Masculino , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Células HEK293 , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas tau/metabolismo , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo , Ubiquitinación/efectos de los fármacos
12.
Front Cell Dev Biol ; 9: 752753, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34746146

RESUMEN

Introduction: Type 2 diabetes mellitus (T2DM) is an independent risk factor of Alzheimer's disease (AD), and populations with mild cognitive impairment (MCI) have high incidence to suffer from AD. Therefore, discerning who may be more vulnerable to MCI, among the increasing T2DM populations, is important for early intervention and eventually decreasing the prevalence rate of AD. This study was to explore whether the change of plasma ß-amyloid (Aß) could be a biomarker to distinguish MCI (T2DM-MCI) from non-MCI (T2DM-nMCI) in T2DM patients. Methods: Eight hundred fifty-two T2DM patients collected from five medical centers were assigned randomly to training and validation cohorts. Plasma Aß, platelet glycogen synthase kinase-3ß (GSK-3ß), apolipoprotein E (ApoE) genotypes, and olfactory and cognitive functions were measured by ELISA, dot blot, RT-PCR, Connecticut Chemosensory Clinical Research Center (CCCRC) olfactory test based on the diluted butanol, and Minimum Mental State Examination (MMSE) test, respectively, and multivariate logistic regression analyses were applied. Results: Elevation of plasma Aß1-42/Aß1-40 is an independent risk factor of MCI in T2DM patients. Although using Aß1-42/Aß1-40 alone only reached an AUC of 0.631 for MCI diagnosis, addition of the elevated Aß1-42/Aß1-40 to our previous model (i.e., activated platelet GSK-3ß, ApoE ε4 genotype, olfactory decline, and aging) significantly increased the discriminating efficiency of T2DM-MCI from T2DM-nMCI, with an AUC of 0.846 (95% CI: 0.794-0.897) to 0.869 (95% CI: 0.822-0.916) in the training cohort and an AUC of 0.848 (95% CI: 0.815-0.882) to 0.867 (95% CI: 0.835-0.899) in the validation cohort, respectively. Conclusion: A combination of the elevated plasma Aß1-42/Aß1-40 with activated platelet GSK-3ß, ApoE ε4 genotype, olfactory decline, and aging could efficiently diagnose MCI in T2DM patients. Further longitudinal studies may consummate the model for early prediction of AD.

13.
Theranostics ; 10(21): 9674-9685, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32863953

RESUMEN

Adult hippocampal neurogenesis (AHN) deficits contribute to the progression of cognitive impairments during accelerated senescence, with the mechanistic causes poorly understood. Glycogen synthase kinase-3ß (GSK-3ß) is a critical regulator in prenatal neurodevelopment. The present study aims to study whether and how GSK-3ß regulates AHN during the accelerated senescence. Methods: AHN and AHN-dependent cognition and GSK-3ß were evaluated in 3- and 6-month senescence-accelerated mice prone 8 (SAM-P8) and senescence resistant 1 (SAM-R1) mice, respectively. GSK-3ß was selectively overexpressed in wild-type mice using adeno-associated virus, or knocked-out by crossbreeding with GSK-3ß floxed mice in the neural stem cells (NSCs) of Nestin-Cre mice, or pharmacologically inhibited with SB216763 in SAM-P8 mice. AHN was evaluated by BrdU-, DCX-staining and retrovirus-labeling. Results: AHN transiently increased at 3-month, but dramatically dropped at 6-month of age in SAM-P8 mice with a simultaneous activation of GSK-3ß at 3-month. Selective overexpression of GSK-3ß in hippocampal NSCs of wildtype mice induced long-term AHN deficits due to an accelerated depletion of NSC pool, although it transiently increased the proliferation and survival of the newborn neurons. Pharmacologically inhibiting GSK-3ß by SB216763 efficiently preserved AHN and improved contextual memory in 6-month SAM-P8 mice, while conditional knock-out of GSK-3ß in NSCs impaired AHN. Conclusion: Early-stage activation of GSK-3ß in NSCs impairs AHN by accelerating the depletion of NSC pool, and pharmacological inhibition of GSK-3ß is efficient to preserve AHN during the accelerated aging. These results reveal novel mechanisms underlying the AHN impairments during accelerated senescence and provide new targets for pro-neurogenic therapies for related diseases.


Asunto(s)
Envejecimiento/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/metabolismo , Neurogénesis/fisiología , Envejecimiento/patología , Animales , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Proteína Doblecortina , Hipocampo/patología , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuronas/metabolismo , Transducción de Señal/fisiología
14.
Biosci Rep ; 40(2)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32043530

RESUMEN

Abnormal aggregation of pathological tau protein is a neuropathological feature of Alzheimer's disease (AD). In the AD patients, the abnormal tau accumulation first appeared in entorhinal cortex (EC) and then propagated to the hippocampus with microglia activation and inflammation, but the mechanism is elusive. Here, we studied the role and mechanisms underlying periphery inflammation on brain tau transmission. By intraperitoneal injection of lipopolysaccharide (LPS) with brain medial entorhinal cortex (MEC)-specific overexpressing P301L human tau (P301L-hTau), we found that both acute and chronic administration of LPS remarkably promoted P301L-hTau transmission from MEC to the hippocampal subsets. Interestingly, the chronic LPS-induced P301L-hTau transmission was still apparent after blocking microglia activation. Further studies demonstrated that LPS disrupted the integrity of blood-brain barrier (BBB) and simultaneous intraperitoneal administration of glucocorticoid (GC) attenuated LPS-promoted P301L-hTau transmission. These data together suggest that a non-microglia-dependent BBB disruption contributes to peripheral LPS-promoted brain P301L-hTau transmission, therefore, maintaining the integrity of BBB can be a novel strategy for preventing pathological tau propagation in AD and other tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Barrera Hematoencefálica/metabolismo , Corteza Entorrinal/metabolismo , Hipocampo/metabolismo , Inflamación/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Conducta Animal , Barrera Hematoencefálica/patología , Cognición , Modelos Animales de Enfermedad , Prueba de Laberinto Elevado , Corteza Entorrinal/patología , Corteza Entorrinal/fisiopatología , Hipocampo/patología , Hipocampo/fisiopatología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Prueba del Laberinto Acuático de Morris , Actividad Motora , Prueba de Campo Abierto , Transporte de Proteínas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteínas tau/genética
15.
J Alzheimers Dis ; 77(1): 241-255, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32804150

RESUMEN

BACKGROUND: Increased tau acetylation at K174, K274, K280, and K281 has been observed in the brains of Alzheimer's disease (AD) patients or in transgenic mice, but the role of acetylation in tau propagation is elusive. OBJECTIVE: To study the effect of tau acetylation in entorhinal cortex on tau transmission and learning and memory. METHODS: Stereotactic brain injection, behavioral test, electrophysiological recording, immunohistochemistry, and immunofluorescence were used. RESULTS: We constructed the hyperacetylation mimics of tau (AAV-Tau-4Q), the non-acetylation tau mutant (AAV-Tau-4R), and the wild-type tau (AAV-Tau-WT). By overexpressing these different tau proteins in the entorhinal cortex (EC) of 2-month-old mice, we found that overexpressing Tau-4Q in EC for 3 or 6 months (to 5 or 8 months of age) neither induces tau propagation to dentate gyrus (DG) nor glial activation in DG, nor spatial memory deficit. However, overexpressing Tau-WT and Tau-4Q in EC for 13.5 months (15.5 months of age) at 2 months promoted tau propagation respectively to granulosa and hilus of DG with glial activation, synaptic dysfunction, and memory deficit, while overexpressing Tau-4R abolished tau propagation with improved cellular pathologies and cognitive functions. Furthermore, overexpressing Tau-4Q in unilateral DG of 2-month-old mice for 8 weeks also promoted its contralateral transmission with glial activation, and mice with tau (Tau-WT, Tau-4Q, and Tau-4R) overexpression in DG showed cognitive deficits compared with the empty vector controls. CONCLUSION: Tau acetylation induces a time-dependent propagation from EC to DG, and only hippocampus but not EC tau accumulation induces cognitive deficits.


Asunto(s)
Disfunción Cognitiva/metabolismo , Corteza Entorrinal/metabolismo , Hipocampo/metabolismo , Proteínas tau/metabolismo , Acetilación , Animales , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Corteza Entorrinal/patología , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas tau/genética
16.
Front Cell Dev Biol ; 8: 633725, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33681188

RESUMEN

Intracellular deposition of hyperphosphorylated tau has been reported in the brain of epilepsy patients, but its contribution to epileptic seizures and the association with spatial cognitive functions remain unclear. Here, we found that repeated optogenetic stimulation of the excitatory neurons in ventral hippocampal CA1 subset could induce a controllable epileptic seizure in mice. Simultaneously, the mice showed spatial learning and memory deficits with a prominently elevated total tau and phospho-tau levels in the brain. Importantly, selective facilitating tau degradation by using a novel designed proteolysis-targeting chimera named C4 could effectively ameliorate the epileptic seizures with remarkable restoration of neuronal firing activities and improvement of spatial learning and memory functions. These results confirm that abnormal tau accumulation plays a pivotal role in the epileptic seizures and the epilepsy-associated spatial memory impairments, which provides new molecular target for the therapeutics.

17.
J Alzheimers Dis ; 73(3): 1105-1123, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31884489

RESUMEN

The three isoforms of 3R-tau are predominantly deposited in neurons bearing neurofibrillary tangles in Alzheimer's disease (AD), while only 3R-tau accumulation has been detected in Pick's disease (PiD), suggesting the involvement of 3R-tau in neurodegeneration. However, both the role and the molecular mechanism of 3R-tau in neurodegeneration are elusive. Here, we co-expressed three isoforms of human wild-type 3R-tau in adult mouse hippocampal to mimic the pathologic tau accumulating observed in PiD patients. We found that co-expressing three 3R-tau isoforms induced hyperphosphorylation and accumulation of tau proteins; simultaneously, the mice showed remarkable neuron death with synapse and memory deficits. Further in vitro and in vivo studies demonstrated that co-expressing 3R-tau isoforms caused oxidative stress evidenced by an increased malondialdehyde, and the decreased superoxide dismutase and glutathione peroxidase; the 3R-tau accumulation also induced significant glial activation and DNA double-strand breaks (DSBs). Notably, the toxic effects of 3R-tau accumulation were efficiently reversed by administration of antioxidants Vitamin E (VitE) and Vitamin C (VitC), respectively. These data reveal that intracellular accumulation of 3R-tau isoforms in adult brain induces significant neuron death and memory deficits with the mechanism involving oxidation-mediated DSBs; and the antioxidants VitE and VitC can efficiently attenuate the toxicities of 3R-tau. Given that no significant cell death has been detected in the currently available wild-type tau-accumulating models, co-expressing 3R-tau isoforms could be a promising model for drug development of tauopathies, such as PiD.


Asunto(s)
Muerte Celular/fisiología , Daño del ADN/fisiología , Trastornos de la Memoria/metabolismo , Isoformas de Proteínas/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Trastornos de la Memoria/genética , Ratones , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/fisiología , Fosforilación , Isoformas de Proteínas/genética , Tauopatías/genética , Tauopatías/patología , Proteínas tau/genética
18.
Transl Neurodegener ; 8: 8, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30867903

RESUMEN

BACKGROUND: Building brain reserves before dementia onset could represent a promising strategy to prevent Alzheimer's disease (AD), while how to initiate early cognitive stimulation is unclear. Given that the immature brain is more sensitive to environmental stimuli and that brain dynamics decrease with ageing, we reasoned that it would be effective to initiate cognitive stimulation against AD as early as the fetal period. METHODS: After conception, maternal AD transgenic mice (3 × Tg AD) were exposed to gestational environment enrichment (GEE) until the day of delivery. The cognitive capacity of the offspring was assessed by the Morris water maze and contextual fear-conditioning tests when the offspring were raised in a standard environment to 7 months of age. Western blotting, immunohistochemistry, real-time PCR, immunoprecipitation, chromatin immunoprecipitation (ChIP) assay, electrophysiology, Golgi staining, activity assays and sandwich ELISA were employed to gain insight into the mechanisms underlying the beneficial effects of GEE on embryos and 7-10-month-old adult offspring. RESULTS: We found that GEE markedly preserved synaptic plasticity and memory capacity with amelioration of hallmark pathologies in 7-10-m-old AD offspring. The beneficial effects of GEE were accompanied by global histone hyperacetylation, including those at bdnf promoter-binding regions, with robust BDNF mRNA and protein expression in both embryo and progeny hippocampus. GEE increased insulin-like growth factor 1 (IGF1) and activated its receptor (IGF1R), which phosphorylates Ca2+/calmodulin-dependent kinase IV (CaMKIV) at tyrosine sites and triggers its nuclear translocation, subsequently upregulating histone acetyltransferase (HAT) and BDNF transcription. The upregulation of IGF1 mimicked the effects of GEE, while IGF1R or HAT inhibition during pregnancy abolished the GEE-induced CaMKIV-dependent histone hyperacetylation and BDNF upregulation. CONCLUSIONS: These findings suggest that activation of IGF1R/CaMKIV/HAT/BDNF signaling by gestational environment enrichment may serve as a promising strategy to delay AD progression.

19.
J Alzheimers Dis ; 63(2): 761-772, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29660945

RESUMEN

The transient receptor potential cation (TRPC) channels are widely expressed in nervous system but their functions remain largely unclear. Here, we found that TRPC1 deletion did not affect learning and memory in physiological conditions, while it aggravated learning and memory deficits induced by amyloid-ß (Aß), the major component of the senile plaques observed in the brains of Alzheimer's disease (AD). Further studies demonstrated that TRPC1 deletion did not affect cell apoptosis in physiological condition, but it exacerbated the Aß-induced cell death in mouse hippocampus. Moreover, the level of TRPC1 was decreased in AD cell and mouse models, and upregulation of TRPC1 decreased Aß levels with attenuation of apoptosis in the cells stably overexpressing amyloid-ß protein precursor (AßPP). Finally, the transmembrane domain of TRPC1 could bind to AßPP and thus decreased Aß production. These findings indicate that loss of TRPC1 exacerbates Aß-induced memory deficit and cell apoptosis, though it does not impair cognitive function or induce cell death in physiological conditions.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apoptosis/fisiología , Trastornos de la Memoria/metabolismo , Fragmentos de Péptidos/metabolismo , Canales Catiónicos TRPC/deficiencia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células HEK293 , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Aprendizaje/fisiología , Masculino , Memoria/fisiología , Trastornos de la Memoria/patología , Ratones de la Cepa 129 , Ratones Noqueados , Dominios Proteicos , Canales Catiónicos TRPC/genética
20.
J Alzheimers Dis ; 66(1): 333-345, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30282353

RESUMEN

Extracellular accumulation of amyloid-ß (Aß) forming senile plaques is one of the hallmark pathologies in Alzheimer's disease (AD), while the mechanisms underlying the neuronal toxic effect of Aß are not fully understood. Here, we found that intracerebroventricular infusion of the aged Aß42 in mice only induces memory deficit at 24 h but not at 7 days. Interestingly, a remarkably increased CREB (cAMP response element-binding protein) Ser133-phosphorylation (pS133-CREB) with microglial activation was detected at 24 h but not at 7 days after Aß infusion. Aß treatment for 24 h increased pS133-CREB level in microglia of the hippocampal non-granular cell layers with remarkably decreased pS133-CREB immunoreactivity in neurons of the hippocampal granular cell layers, including CA1, CA3, and DG subsets. Inhibition of microglia activation by minocycline or CREB phosphorylation by H89, an inhibitor of protein kinase A (PKA), abolished Aß-induced microglia CREB hyperphosphorylation with restoration of neuronal function and attenuation of inflammatory response, i.e., reduced levels of interleukin-6 (IL6) and pCREB binding of matrix metalloproteinase-9 (MMP9) DNA. Finally, treatment of the primary hippocampal neurons with Aß-potentiated microglia media decreased neuronal GluN1 and GluA2 levels, while simultaneous inhibition of PKA restored the levels. These novel findings reveal that intracerebroventricular infusion of Aß only induces transient memory deficit in mice and the molecular mechanisms involve a stimulated microglial CREB phosphorylation.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/toxicidad , Animales , Células Cultivadas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Infusiones Intraventriculares , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA