Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.167
Filtrer
1.
CNS Neurosci Ther ; 30(7): e14818, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38946682

RÉSUMÉ

Glycogen synthase kinase-3 (GSK3), consisting of GSK3α and GSK3ß subtypes, is a complex protein kinase that regulates numerous substrates. Research has observed increased GSK3 expression in the brains of Alzheimer's disease (AD) patients and models. AD is a neurodegenerative disorder with diverse pathogenesis and notable cognitive impairments, characterized by Aß aggregation and excessive tau phosphorylation. This article provides an overview of GSK3's structure and regulation, extensively analyzing its relationship with AD factors. GSK3 overactivation disrupts neural growth, development, and function. It directly promotes tau phosphorylation, regulates amyloid precursor protein (APP) cleavage, leading to Aß formation, and directly or indirectly triggers neuroinflammation and oxidative damage. We also summarize preclinical research highlighting the inhibition of GSK3 activity as a primary therapeutic approach for AD. Finally, pending issues like the lack of highly specific and affinity-driven GSK3 inhibitors, are raised and expected to be addressed in future research. In conclusion, GSK3 represents a target in AD treatment, filled with hope, challenges, opportunities, and obstacles.


Sujet(s)
Maladie d'Alzheimer , Glycogen Synthase Kinase 3 , Animaux , Humains , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/enzymologie , Précurseur de la protéine bêta-amyloïde/métabolisme , Glycogen Synthase Kinase 3/antagonistes et inhibiteurs , Glycogen Synthase Kinase 3/métabolisme , Protéines tau/métabolisme , Protéines tau/antagonistes et inhibiteurs
2.
Mol Neurobiol ; 61(7): 4834-4853, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38135855

RÉSUMÉ

With the aging of the population, treatment of conditions emerging in old age, such as neurodegenerative disorders, has become a major medical challenge. Of these, Alzheimer's disease, leading to cognitive dysfunction, is of particular interest. Neuronal loss plays an important role in the pathophysiology of this condition, and over the years, a great effort has been made to determine the role of various factors in this process. Unfortunately, until now, the exact pathomechanism of this condition remains unknown. However, the most popular theories associate AD with abnormalities in the Tau and ß-amyloid (Aß) proteins, which lead to their deposition and result in neuronal death. Neurons, like all cells, die in a variety of ways, among which pyroptosis, apoptosis, and necroptosis are associated with the activation of various caspases. It is worth mentioning that Tau and Aß proteins are considered to be one of the caspase activators, leading to cell death. Moreover, the protease activity of caspases influences both of the previously mentioned proteins, Tau and Aß, converting them into more toxic derivatives. Due to the variety of ways caspases impact the development of AD, drugs targeting caspases could potentially be useful in the treatment of this condition. Therefore, there is a constant need to search for novel caspase inhibitors and evaluate them in preclinical and clinical trials.


Sujet(s)
Maladie d'Alzheimer , Caspases , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/métabolisme , Humains , Caspases/métabolisme , Animaux , Protéines tau/métabolisme , Peptides bêta-amyloïdes/métabolisme , Activation enzymatique , Inhibiteurs des caspases/usage thérapeutique , Inhibiteurs des caspases/pharmacologie
3.
J Biol Chem ; 299(12): 105411, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37918804

RÉSUMÉ

O-GlcNAc is a common modification found on nuclear and cytoplasmic proteins. Determining the catalytic mechanism of the enzyme O-GlcNAcase (OGA), which removes O-GlcNAc from proteins, enabled the creation of potent and selective inhibitors of this regulatory enzyme. Such inhibitors have served as important tools in helping to uncover the cellular and organismal physiological roles of this modification. In addition, OGA inhibitors have been important for defining the augmentation of O-GlcNAc as a promising disease-modifying approach to combat several neurodegenerative diseases including both Alzheimer's disease and Parkinson's disease. These studies have led to development and optimization of OGA inhibitors for clinical application. These compounds have been shown to be well tolerated in early clinical studies and are steadily advancing into the clinic. Despite these advances, the mechanisms by which O-GlcNAc protects against these various types of neurodegeneration are a topic of continuing interest since improved insight may enable the creation of more targeted strategies to modulate O-GlcNAc for therapeutic benefit. Relevant pathways on which O-GlcNAc has been found to exert beneficial effects include autophagy, necroptosis, and processing of the amyloid precursor protein. More recently, the development and application of chemical methods enabling the synthesis of homogenous proteins have clarified the biochemical effects of O-GlcNAc on protein aggregation and uncovered new roles for O-GlcNAc in heat shock response. Here, we discuss the features of O-GlcNAc in neurodegenerative diseases, the application of inhibitors to identify the roles of this modification, and the biochemical effects of O-GlcNAc on proteins and pathways associated with neurodegeneration.


Sujet(s)
Maladie d'Alzheimer , N-acetylglucosaminyltransferase , Maladie de Parkinson , Humains , Acétyl-glucosamine/métabolisme , Maladie d'Alzheimer/enzymologie , Précurseur de la protéine bêta-amyloïde/métabolisme , beta-N-Acetylhexosaminidases/génétique , N-acetylglucosaminyltransferase/antagonistes et inhibiteurs , N-acetylglucosaminyltransferase/métabolisme , Maladie de Parkinson/enzymologie , Maturation post-traductionnelle des protéines , Antienzymes/pharmacologie
4.
J Biol Chem ; 299(6): 104794, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37164155

RÉSUMÉ

Clinical development of γ-secretases, a family of intramembrane cleaving proteases, as therapeutic targets for a variety of disorders including cancer and Alzheimer's disease was aborted because of serious mechanism-based side effects in the phase III trials of unselective inhibitors. Selective inhibition of specific γ-secretase complexes, containing either PSEN1 or PSEN2 as the catalytic subunit and APH1A or APH1B as supporting subunits, does provide a feasible therapeutic window in preclinical models of these disorders. We explore here the pharmacophoric features required for PSEN1 versus PSEN2 selective inhibition. We synthesized a series of brain penetrant 2-azabicyclo[2,2,2]octane sulfonamides and identified a compound with low nanomolar potency and high selectivity (>250-fold) toward the PSEN1-APH1B subcomplex versus PSEN2 subcomplexes. We used modeling and site-directed mutagenesis to identify critical amino acids along the entry part of this inhibitor into the catalytic site of PSEN1. Specific targeting one of the different γ-secretase complexes might provide safer drugs in the future.


Sujet(s)
Amyloid precursor protein secretases , Complexes multiprotéiques , Préséniline-1 , Sulfonamides , Humains , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/métabolisme , Amyloid precursor protein secretases/antagonistes et inhibiteurs , Amyloid precursor protein secretases/métabolisme , Préséniline-1/antagonistes et inhibiteurs , Préséniline-1/métabolisme , Complexes multiprotéiques/antagonistes et inhibiteurs , Complexes multiprotéiques/métabolisme , Sulfonamides/pharmacologie , Spécificité du substrat , Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Tumeurs/métabolisme
5.
Eur J Med Chem ; 245(Pt 1): 114894, 2023 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-36343411

RÉSUMÉ

Despite innumerable efforts to develop effective therapeutics, it is difficult to achieve breakthrough treatments for Alzheimer's disease (AD), and the main reason is probably the absence of a clear target. Here, we reveal c-Jun N-terminal kinase 3 (JNK3), a protein kinase explicitly expressed in the brain and involved in neuronal apoptosis, with a view toward providing effective treatment for AD. For many years, we have worked on JNK3 inhibitors and have discovered 2-aryl-1-pyrimidinyl-1H-imidazole-5-yl acetonitrile-based JNK3 inhibitors with superb potency (IC50 < 1.0 nM) and excellent selectivity over other protein kinases including isoforms JNK1 (>300 fold) and JNK2 (∼10 fold). Based on in vitro biological activity and DMPK properties, the lead compounds were selected for further in vivo studies. We confirmed that repeat administration of JNK3 inhibitors improved cognitive memory in APP/PS1 and the 3xTg mouse model. Overall, our results show that JNK3 could be a potential target protein for AD.


Sujet(s)
Maladie d'Alzheimer , Imidazoles , Mitogen-Activated Protein Kinase 10 , Inhibiteurs de protéines kinases , Animaux , Souris , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/enzymologie , Apoptose/effets des médicaments et des substances chimiques , Imidazoles/composition chimique , Imidazoles/pharmacologie , Imidazoles/usage thérapeutique , Mitogen-Activated Protein Kinase 10/antagonistes et inhibiteurs , Isoformes de protéines/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Modèles animaux de maladie humaine
6.
Eur J Med Chem ; 244: 114837, 2022 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-36265279

RÉSUMÉ

The toxic pyroglutamate form of amyloid-ß (pE-Aß) is important for the pathogenesis of early Alzheimer's disease (AD); therefore, reducing pE-Aß by inhibiting glutaminyl cyclase (QC) provides a promising strategy for developing disease-modifying AD drugs. In this study, potent and selective QC inhibitors with desirable drug-like properties were discovered by replacing the 3,4-dimethoxyphenyl group in a QC inhibitor with a bioisosteric indazole surrogate. Among them, 3-methylindazole-6-yl and 3-methylindazole-5-yl derivatives with an N-cyclohexylurea were identified as highly potent inhibitors with IC50 values of 3.2 nM and 2.3 nM, respectively, both of which were approximately 10-fold more potent than varoglutamstat. In addition, the three inhibitors significantly reduced pE-Aß3-40 levels in an acute animal model after intracerebroventricular (icv) injection and were selective for hQC. Further in vitro pharmacokinetic and toxicity studies, including those investigating cytotoxicity, hERG inhibition, blood-brain barrier (BBB) permeability and metabolic stability, indicated that N-(3-methylindazole-6-yl)-N'-(cyclohexyl)urea derivative exhibited the most promising efficacy, selectivity and drug-like profile; thus, it was evaluated for its in vivo efficacy in an AD model.


Sujet(s)
Maladie d'Alzheimer , Aminoacyltransferases , Découverte de médicament , Indazoles , Animaux , Humains , Maladie d'Alzheimer/enzymologie , Aminoacyltransferases/antagonistes et inhibiteurs , Aminoacyltransferases/composition chimique , Peptides bêta-amyloïdes/métabolisme , Indazoles/composition chimique , Indazoles/pharmacologie
7.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-35806318

RÉSUMÉ

Increasing evidence implicates endothelial dysfunction in the pathogenesis of Alzheimer's disease (AD). Nitric oxide (NO) derived from endothelial NO synthase (eNOS) is essential in maintaining cerebrovascular function and can modulate the production and clearance of amyloid beta (Aß). APPswe/PSdE1 (APP/PS1) mice display age-related Aß accumulation and memory deficits. In order to make the model more clinically relevant with an element of endothelial dysfunction, we generated APP/PS1/eNOS+/- mice by crossing complete eNOS deficient (eNOS-/-) mice and APP/PS1 mice. APP/PS1/eNOS+/- mice at 8 months of age displayed a more severe spatial working memory deficit relative to age-matched APP/PS1 mice. Moreover, immunohistochemistry and immunoblotting revealed significantly increased Aß plaque load in the brains of APP/PS1/eNOS+/- mice, concomitant with upregulated BACE-1 (hence increased Aß production), downregulated insulin-degrading enzyme (hence reduced Aß clearance) and increased immunoreactivity and expression of microglia. The present study, for the first time, demonstrated that partial eNOS deficiency exacerbated behavioral dysfunction, Aß brain deposition, and microglial pathology in APP/PS1 mice, further implicating endothelial dysfunction in the pathogenesis of AD. The present findings also provide the scientific basis for developing preventive and/or therapeutic strategies by targeting endothelial dysfunction.


Sujet(s)
Maladie d'Alzheimer , Dysfonctionnement cognitif , Nitric oxide synthase type III , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/psychologie , Peptides bêta-amyloïdes/génétique , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Animaux , Dysfonctionnement cognitif/enzymologie , Dysfonctionnement cognitif/génétique , Dysfonctionnement cognitif/métabolisme , Modèles animaux de maladie humaine , Troubles de la mémoire/génétique , Troubles de la mémoire/métabolisme , Souris , Souris transgéniques , Nitric oxide synthase type III/déficit , Nitric oxide synthase type III/génétique , Nitric oxide synthase type III/métabolisme , Plaque amyloïde/métabolisme , Préséniline-1/métabolisme
8.
Biochem Pharmacol ; 201: 115071, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35525328

RÉSUMÉ

Despite large investments by industry and governments, no disease-modifying medications for the treatment of patients with Alzheimer's disease (AD) have been found. The failures of various clinical trials indicate the need for a more in-depth understanding of the pathophysiology of AD and for innovative therapeutic strategies for its treatment. Here, we review the rational for targeting IP3 signaling, cytosolic calcium dysregulation, phosphodiesterases (PDEs), and secondary messengers like cGMP and cAMP, as well as their correlations with the pathophysiology of AD. Various drugs targeting these signaling cascades are still in pre-clinical and clinical trials which support the ideas presented in this article. Further, we describe different molecular mechanisms and medications currently being used in various pre-clinical and clinical trials involving IP3/Ca+2 signaling. We also highlight various isoforms, as well as the functions and pharmacology of the PDEs broadly expressed in different parts of the brain and attempt to unravel the potential benefits of PDE inhibitors for use as novel medications to alleviate the pathogenesis of AD.


Sujet(s)
Maladie d'Alzheimer , Signalisation calcique , Récepteurs à l'inositol 1,4,5-triphosphate , Phosphodiesterases , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Signalisation calcique/effets des médicaments et des substances chimiques , GMP cyclique/métabolisme , Humains , Récepteurs à l'inositol 1,4,5-triphosphate/métabolisme , Thérapie moléculaire ciblée , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Phosphodiesterases/métabolisme , Transduction du signal
9.
Neurobiol Dis ; 169: 105737, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35452786

RÉSUMÉ

Altered mitochondrial DNA (mtDNA) occurs in neurodegenerative disorders like Alzheimer's disease (AD); how mtDNA synthesis is linked to neurodegeneration is poorly understood. We previously discovered Nutrient-induced Mitochondrial Activity (NiMA), an inter-organelle signaling pathway where nutrient-stimulated lysosomal mTORC1 activity regulates mtDNA replication in neurons by a mechanism sensitive to amyloid-ß oligomers (AßOs), a primary factor in AD pathogenesis (Norambuena et al., 2018). Using 5-ethynyl-2'-deoxyuridine (EdU) incorporation into mtDNA of cultured neurons, along with photoacoustic and mitochondrial metabolic imaging of cultured neurons and mouse brains, we show these effects being mediated by mTORC1-catalyzed T40 phosphorylation of superoxide dismutase 1 (SOD1). Mechanistically, tau, another key factor in AD pathogenesis and other tauopathies, reduced the lysosomal content of the tuberous sclerosis complex (TSC), thereby increasing NiMA and suppressing SOD1 activity and mtDNA synthesis. AßOs inhibited these actions. Dysregulation of mtDNA synthesis was observed in fibroblasts derived from tuberous sclerosis (TS) patients, who lack functional TSC and elevated SOD1 activity was also observed in human AD brain. Together, these findings imply that tau and SOD1 couple nutrient availability to mtDNA replication, linking mitochondrial dysfunction to AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Superoxide dismutase-1 , Complexe de la sclérose tubéreuse , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/génétique , Peptides bêta-amyloïdes/métabolisme , Animaux , ADN mitochondrial/génétique , ADN mitochondrial/métabolisme , Humains , Lysosomes/génétique , Lysosomes/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Souris , Mitochondries/métabolisme , Superoxide dismutase-1/génétique , Superoxide dismutase-1/métabolisme , Complexe de la sclérose tubéreuse/enzymologie , Complexe de la sclérose tubéreuse/génétique
10.
J Biol Chem ; 298(6): 101911, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35398353

RÉSUMÉ

Neurotoxic amyloid ß-peptides are thought to be a causative agent of Alzheimer's disease in humans. The production of amyloid ß-peptides from amyloid precursor protein (APP) could be diminished by enhancing α-processing; however, the physical interactions between APP and α-secretases are not well understood. In this study, we employed super-resolution light microscopy to examine in cell-free plasma membranes the abundance and association of APP and α-secretases ADAM10 (a disintegrin and metalloproteinase) and ADAM17. We found that both secretase molecules localize similarly closely to APP (within ≤50 nm). However, when cross-linking APP with antibodies directed against the GFP tag of APP, in confocal microscopy, we observed that only ADAM10 coaggregated with APP. Furthermore, we mapped the involved protein domain by using APP variants with an exchanged transmembrane segment or lacking cytoplasmic/extracellular domains. We identified that the transmembrane domain of APP is required for association with α-secretases and, as analyzed by Western blot, for α-processing. We propose that the transmembrane domain of APP interacts either directly or indirectly with ADAM10, but not with ADAM17, explaining the dominant role of ADAM10 in α-processing of APP. Further understanding of this interaction may facilitate the development of a therapeutic strategy based on promoting APP cleavage by α-secretases.


Sujet(s)
Protéine ADAM10 , Maladie d'Alzheimer , Amyloid precursor protein secretases , Précurseur de la protéine bêta-amyloïde , Protéine ADAM10/génétique , Protéine ADAM10/métabolisme , Protéine ADAM17/génétique , Protéine ADAM17/métabolisme , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/génétique , Amyloid precursor protein secretases/génétique , Amyloid precursor protein secretases/métabolisme , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Humains , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Domaines protéiques
11.
Int J Biol Sci ; 18(2): 693-706, 2022.
Article de Anglais | MEDLINE | ID: mdl-35002518

RÉSUMÉ

The aggregation of amyloid-ß (Aß) peptides into oligomers and fibrils is a key pathological feature of Alzheimer's disease (AD). An increasing amount of evidence suggests that oligomeric Aß might be the major culprit responsible for various neuropathological changes in AD. Death-associated protein kinase 1 (DAPK1) is abnormally elevated in brains of AD patients and plays an important role in modulating tau homeostasis by regulating prolyl isomerase Pin1 phosphorylation. However, it remains elusive whether and how Aß species influence the function of DAPK1, and whether this may further affect the function and phosphorylation of tau in neurons. Herein, we demonstrated that Aß aggregates (both oligomers and fibrils) prepared from synthetic Aß42 peptides were able to upregulate DAPK1 protein levels and thereby its function through heat shock protein 90 (HSP90)-mediated protein stabilization. DAPK1 activation not only caused neuronal apoptosis, but also phosphorylated Pin1 at the Ser71 residue, leading to tau accumulation and phosphorylation at multiple AD-related sites in primary neurons. Both DAPK1 knockout (KO) and the application of a specific DAPK1 inhibitor could effectively protect primary neurons against Aß aggregate-induced cell death and tau dysregulation, corroborating the critical role of DAPK1 in mediating Aß aggregation-induced neuronal damage. Our study suggests a mechanistic link between Aß oligomerization and tau hyperphosphorylation mediated by DAPK1, and supports the role of DAPK1 as a promising target for early intervention in AD.


Sujet(s)
Maladie d'Alzheimer/enzymologie , Peptides bêta-amyloïdes/métabolisme , Encéphale/enzymologie , Death-associated protein kinases/métabolisme , Neurones/enzymologie , Fragments peptidiques/métabolisme , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/génétique , Animaux , Apoptose/génétique , Encéphale/anatomopathologie , Death-associated protein kinases/déficit , Death-associated protein kinases/génétique , Modèles animaux de maladie humaine , Souris , Souris de lignée C57BL , Souris knockout , NIMA-interacting peptidylprolyl isomerase/métabolisme , Neurones/anatomopathologie , Fragments peptidiques/génétique , Phosphorylation , Protéines tau/génétique , Protéines tau/métabolisme
12.
Anal Chem ; 94(2): 1491-1497, 2022 01 18.
Article de Anglais | MEDLINE | ID: mdl-34985875

RÉSUMÉ

An amyloid-beta peptide (Aß) is generally believed to be a pathological marker of Alzheimer's disease (AD), but it is still of great significance to explore the upstream and downstream relationship of Aß in AD. It is previously reported that c-Abl, a nonreceptor tyrosine kinase, can be activated by Aß, but the interaction between Aß and c-Abl is still unknown. Herein, an extended-gate field-effect transistor (EG-FET)-based sensor has been developed to monitor the level of c-Abl with high sensitivity and selectivity. Our peptide-functionalized EG-FET sensor as the signal transducer can follow c-Abl activity with electron transfer by its specific phosphorylation. The sensor presents a good linear correlation over c-Abl concentrations of 1 pg/mL to 3.05 µg/mL. The sensor was successfully applied to quantify c-Abl activity in the brain tissue of AD transgenic mice, and the interaction between c-Abl and Aß in AD mice was explored by administering the c-Abl inhibitor (imatinib) and the agonist (DPH). Our work is expected to provide an important reference for early diagnosis and treatment of AD.


Sujet(s)
Maladie d'Alzheimer , Techniques de biocapteur , Protéines proto-oncogènes c-abl , Transistors électroniques , Maladie d'Alzheimer/diagnostic , Maladie d'Alzheimer/enzymologie , Peptides bêta-amyloïdes/métabolisme , Animaux , Marqueurs biologiques/analyse , Marqueurs biologiques/métabolisme , Encéphale/métabolisme , Modèles animaux de maladie humaine , Souris , Souris transgéniques , Phosphorylation , Protéines proto-oncogènes c-abl/analyse , Protéines proto-oncogènes c-abl/métabolisme
13.
J Biochem ; 170(6): 729-738, 2022 Jan 07.
Article de Anglais | MEDLINE | ID: mdl-34523681

RÉSUMÉ

Lemur tail kinase 1 (LMTK1), previously called apoptosis-associated tyrosine kinase (AATYK), is an endosomal Ser/Thr kinase. We recently reported that LMTK1 regulates axon outgrowth, dendrite arborization and spine formation via Rab11-mediated vesicle transport. Rab11, a small GTPase regulating recycling endosome trafficking, is shown to be associated with late-onset Alzheimer's disease (LOAD). In fact, genome-wide association studies identified many proteins regulating vesicle transport as risk factors for LOAD. Furthermore, LMTK1 has been reported to be a risk factor for frontotemporal dementia. Then, we hypothesized that LMTK1 contributes to AD development through vesicle transport and examined the effect of LMTK1 on the cellular localization of AD-related proteins, amyloid precursor protein (APP) and ß-site APP cleaving enzyme 1 (BACE1). The ß-cleavage of APP by BACE1 is the initial and rate-limiting step in Aß generation. We found that LMTK1 accumulated BACE1, but not APP, to the perinuclear endosomal compartment, whereas the kinase-negative(kn) mutant of LMTK1A did not. The ß-C-terminal fragment was prone to increase under overexpression of LMTK1A kn. Moreover, the expression level of LMTK1A was reduced in AD brains. These results suggest the possibility that LMTK1 is involved in AD development through the regulation of the proper endosomal localization of BACE1.


Sujet(s)
Maladie d'Alzheimer/enzymologie , Amyloid precursor protein secretases/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Aspartic acid endopeptidases/métabolisme , Endosomes/enzymologie , Protein-tyrosine kinases/métabolisme , Maladie d'Alzheimer/génétique , Amyloid precursor protein secretases/génétique , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Animaux , Protéines régulatrices de l'apoptose/génétique , Aspartic acid endopeptidases/génétique , Cellules CHO , Cellules COS , Chlorocebus aethiops , Cricetulus , Endosomes/génétique , Cellules HEK293 , Humains , Protein-tyrosine kinases/génétique
14.
Biochim Biophys Acta Mol Cell Res ; 1869(3): 119164, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34699873

RÉSUMÉ

Alzheimer's disease (AD) is the most common form of dementia, however incurable so far. It is widely accepted that aggregated amyloid ß (Aß) peptides play a crucial role for the pathogenesis of AD, as they cause neurotoxicity and deposit as so-called Aß plaques in AD patient brains. Aß peptides derive from the amyloid precursor protein (APP) upon consecutive cleavage at the ß- and γ-secretase site. Hence, mutations in the APP gene are often associated with autosomal dominant inherited AD. Almost thirty years ago, two mutations at the ß-secretase site were observed in two Swedish families (termed Swedish APP (APPswe) mutations), which led to early-onset AD. Consequently, APPswe was established in almost every common AD mouse model, as it contributes to early Aß plaque formation and cognitive impairments. Analyzing these APPswe-based mouse models, the aspartyl protease BACE1 has been evolving as the prominent ß-secretase responsible for Aß release in AD and as the most important therapeutic target for AD treatment. However, with respect to ß-secretase processing, the very rare occurring APPswe variant substantially differs from wild-type APP. BACE1 dominates APPswe processing resulting in the release of Aß1-x, whereas N-terminally truncated Aß forms are scarcely generated. However, these N-terminally truncated Aß species such as Aß2-x, Aß3-x and Aß4-x are elevated in AD patient brains and exhibit an increased potential to aggregate compared to Aß1-x peptides. Proteases such as meprin ß, cathepsin B and ADAMTS4 were identified as alternative ß-secretases being capable of generating these N-terminally truncated Aß species from wild-type APP. However, neither meprin ß nor cathepsin B are capable of generating N-terminally truncated Aß peptides from APPswe. Hence, the role of BACE1 for the Aß formation during AD might be overrepresented through the excessive use of APPswe mouse models. In this review we critically discuss the consideration of BACE1 as the most promising therapeutic target. Shifting the focus of AD research towards alternative ß secretases might unveil promising alternatives to BACE1 inhibitors constantly failing in clinical trials due to ineffectiveness and harmful side effects.


Sujet(s)
Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/anatomopathologie , Amyloid precursor protein secretases/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Modèles animaux de maladie humaine , Amyloid precursor protein secretases/génétique , Animaux , Humains , Souris transgéniques , Suède
15.
Cell Rep ; 37(10): 110102, 2021 12 07.
Article de Anglais | MEDLINE | ID: mdl-34879266

RÉSUMÉ

Toxic amyloid beta (Aß) species cause synaptic dysfunction and neurotoxicity in Alzheimer's disease (AD). As of yet, however, there are no reported regulators for gamma-secretase, which links a risky environment to amyloid accumulation in AD. Here, we report that pyruvate kinase M2 (PKM2) is a positive regulator of gamma-secretase under hypoxia. From a genome-wide functional screen, we identify PKM2 as a gamma-secretase activator that is highly expressed in the brains of both patients and murine models with AD. PKM2 regulates Aß production and the amount of active gamma-secretase complex by changing the gene expression of aph-1 homolog. Hypoxia induces PKM2 expression, thereby promoting gamma-secretase activity. Moreover, transgenic expression of PKM2 in 3xTg AD model mice enhances hippocampal production of Aß and exacerbates the impairment of spatial and recognition memory. Taken together, these findings indicate that PKM2 is an important gamma-secretase regulator that promotes Aß production and memory impairment under hypoxia.


Sujet(s)
Maladie d'Alzheimer/enzymologie , Comportement animal , Encéphale/enzymologie , Endopeptidases/métabolisme , Protéines membranaires/métabolisme , Mémoire , Pyruvate kinase/métabolisme , Sujet âgé , Sujet âgé de 80 ans ou plus , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/physiopathologie , Maladie d'Alzheimer/psychologie , Peptides bêta-amyloïdes/métabolisme , Animaux , Encéphale/physiopathologie , Protéines de transport/génétique , Protéines de transport/métabolisme , Études cas-témoins , Bases de données génétiques , Modèles animaux de maladie humaine , Endopeptidases/génétique , Femelle , Régulation de l'expression des gènes codant pour des enzymes , Humains , Mâle , Protéines membranaires/génétique , Souris de lignée C57BL , Souris transgéniques , Adulte d'âge moyen , Pyruvate kinase/génétique , , Transduction du signal , Mémoire spatiale , Hormones thyroïdiennes/génétique , Hormones thyroïdiennes/métabolisme , Transcription génétique ,
16.
Bioengineered ; 12(2): 12678-12690, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34818971

RÉSUMÉ

Alzheimer's disease (AD) is a progressive neurodegenerative disease. Multiple reports have elucidated that microRNAs are promising biomarkers for AD diagnosis and treatment. Herein, the effect of miR-191-5p on microglial cell injury and the underlying mechanism were explored. APP/PS1 transgenic mice were utilized to establish mouse model of AD. Amyloid-ß protein 1-42 (Aß1-42)-treated microglia were applied to establish in vitro cell model of AD. MiR-191-5p expression in hippocampus and microglia was measured by reverse transcription quantitative polymerase chain reaction. The viability and apoptosis of microglia were evaluated by Cell Counting Kit-8 assays and flow cytometry analyses, respectively. The binding relationship between miR-191-5p and its downstream target mitogen-activated protein kinase kinase kinase 12 (Map3k12) was determined by luciferase reporter assays. Pathological degeneration of hippocampus was tested using hematoxylin-eosin staining and Nissl staining. Aß expression in hippocampus was examined via immunohistochemistry. In this study, miR-191-5p was downregulated in Aß1-42-stimulated microglia and hippocampal tissues of APP/PS1 mice. MiR-191-5p overexpression facilitated cell viability and inhibited apoptosis rate of Aß1-42-treated microglia. Mechanically, miR-191-5p targeted Map3k12 3'-untranslated region to downregulate Map3k12 expression. MiR-191-5p inhibited Aß1-42-induced microglial cell injury and inactivated the MAPK signaling by downregulating Map3k12. Overall, miR-191-5p alleviated Aß1-42-induced microglia cell injury by targeting Map3k12 to inhibit the MAPK signaling pathway in microglia.


Sujet(s)
Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/génétique , MAP Kinase Kinase Kinases/métabolisme , Système de signalisation des MAP kinases , Microglie/enzymologie , Microglie/anatomopathologie , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/toxicité , Animaux , Régulation négative/génétique , Hippocampe/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL , Souris transgéniques , Microglie/effets des médicaments et des substances chimiques , Fragments peptidiques/toxicité
17.
Int J Mol Sci ; 22(20)2021 Oct 17.
Article de Anglais | MEDLINE | ID: mdl-34681851

RÉSUMÉ

Alzheimer's Disease (AD) is the most common neurodegenerative disorder in our society, as the population ages, its incidence is expected to increase in the coming decades. The etiopathology of this disease still remains largely unclear, probably because of the highly complex and multifactorial nature of AD. However, the presence of mitochondrial dysfunction has been broadly described in AD neurons and other cellular populations within the brain, in a wide variety of models and organisms, including post-mortem humans. Mitochondria are complex organelles that play a crucial role in a wide range of cellular processes, including bioenergetics. In fact, in mammals, including humans, the main source of cellular ATP is the oxidative phosphorylation (OXPHOS), a process that occurs in the mitochondrial electron transfer chain (ETC). The last enzyme of the ETC, and therefore the ulterior generator of ATP, is the ATP synthase. Interestingly, in mammalian cells, the ATP synthase can also degrade ATP under certain conditions (ATPase), which further illustrates the crucial role of this enzyme in the regulation of cellular bioenergetics and metabolism. In this collaborative review, we aim to summarize the knowledge of the presence of dysregulated ATP synthase, and of other components of mammalian mitochondrial bioenergetics, as an early event in AD. This dysregulation can act as a trigger of the dysfunction of the organelle, which is a clear component in the etiopathology of AD. Consequently, the pharmacological modulation of the ATP synthase could be a potential strategy to prevent mitochondrial dysfunction in AD.


Sujet(s)
Maladie d'Alzheimer/métabolisme , Mitochondries/métabolisme , Mitochondrial Proton-Translocating ATPases/métabolisme , Neurones/métabolisme , Maladie d'Alzheimer/enzymologie , Animaux , Métabolisme énergétique , Humains , Phosphorylation oxydative
18.
Biomed Pharmacother ; 144: 112271, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34619494

RÉSUMÉ

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and cognitive impairment. ß-Amyloid (Aß) is widely accepted as the main neurotoxin that triggers mitochondrial-associated oxidative stress, leading to neuronal death in AD. Following our preliminary research on the neuroprotective effects of the brown alga Sargassum serratifolium, its major compounds, including sargaquinoic acid, sargahydroquinoic acid (SHQA), and sargachromenol, were investigated to elucidate the antioxidant and anti-apoptotic properties of Aß25-35-stimulated PC12 cells. SHQA exhibited the most potent effect on Aß-induced mitochondrial-associated oxidative stress and apoptosis. In addition, the compound enhanced the expression and translocation of nuclear factor-E2-related factor 2 (Nrf2), while reducing the expression of cytoplasmic Kelch-like ECH-associated protein 1 (Keap1). Furthermore, the compound upregulated the expression of Nrf2-regulated antioxidant enzymes, including HO-1, NQO1, GCLc, GCLm, and TrxR1. Co-treatment with SHQA and LY294002, a specific PI3K inhibitor, inhibited nuclear Nrf2 expression and Akt phosphorylation, demonstrating that SHQA-mediated Nrf2 activation was directly associated with the PI3K/Akt signaling pathway. Mechanistic studies indicate that activation of the PI3K/Akt/Nrf2 pathway is the molecular basis for the neuroprotective effects of SHQA. In silico docking simulation revealed that SHQA established specific interactions with the key amino acid residues of PI3K, Akt, and Nrf2-Keap1 via hydrogen bonding and van der Waals interactions, which may affect the biological capacities of target markers. Overall, this is the first report of this novel mechanism of SHQA as a Nrf2 activator against Aß-mediated oxidative damage, suggesting that the compound might be a potential agent for the prevention of AD.


Sujet(s)
Alcènes/pharmacologie , Maladie d'Alzheimer/traitement médicamenteux , Peptides bêta-amyloïdes/toxicité , Antioxydants/pharmacologie , Benzoquinones/pharmacologie , Facteur-2 apparenté à NF-E2/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Fragments peptidiques/toxicité , Phosphatidylinositol 3-kinase/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/anatomopathologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/enzymologie , Mitochondries/anatomopathologie , Simulation de docking moléculaire , Neurones/enzymologie , Neurones/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Cellules PC12 , Phosphorylation , Rats , Transduction du signal
19.
Int Immunopharmacol ; 100: 108083, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34478946

RÉSUMÉ

Alzheimer's disease (AD) is classified pathologically as a progressive neurological disorder associated with memory decline. The study was designed to assess the underlying molecular signaling involved in the neuroprotective effect of the 2-(hydroxyl-(2-nitrophenyl)methyl)cyclopentanone (2NCP) as a novel therapeutic agent for AD. In this connection, in vitro cholinesterases inhibitory and antioxidant activities were investigated. In vivo studies were carried out on a well-known 5xFAD mice model in different behavioural models such as light/dark box,balance beam, rotarod, elevated plus maze (EPM),novel object recognition (NOR), paddling Y-maze, and Morris water maze (MWM) tests. Hippocampus (HC) and frontal cortex (FC) homogenates were examined for acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) activities, 1,1-diphenyl-2-picrylhydrazyl (DPPH) free radicals, glutathione S-transferase (GST), glutathione (GSH), and catalase. Further, we examined the expression of inflammatory cytokines and Nrf2 in the HC and FC through RT-PCR. Computational studies were conducted to predict the binding mode of the 2NCP with target sites of nuclear factor-κB (NF-κB) and cholinesterases. The findings of in vitro assays revealed that the IC50 values of the 2NCP against AChE and BChE were 17 and 23 µg/ml respectively. DPPH antioxidant assay displayed an IC50 value for the 2NCP was 62 µg/ml. Whereas, theex vivo study depicted that the activities of AChE and BChEwere significantly reduced. Moreover, free radicals load, GSH level, catalase and GST activities were significantly declined. Furthermore, in vivostudies showed that the 2NCP treated animals exhibited gradual memory improvement and improved motor functions. RT-PCR study revealed that mRNA levels of the inflammatory mediators (IL-1ß, IL-6, TNF-α) were significantly reduced, while the expression of antioxidant Nrf2 was significantly increased.The molecular docking studies further confirmed that the 2NCP showed excellent binding affinities for NF-κB and cholinesterases. Taken together, the 2NCP improves spatial memory and learning, short- and long-term memory,markedly inhibits cholinesterases, reduced neuroinflammation, and mitigated oxidative stress in the 5xFAD mice; hence the 2NCP may be a potential candidate for the management of AD.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Anti-inflammatoires/pharmacologie , Antioxydants/pharmacologie , Comportement animal/effets des médicaments et des substances chimiques , Encéphale/effets des médicaments et des substances chimiques , Butyrylcholine esterase/métabolisme , Anticholinestérasiques/pharmacologie , Maladies neuro-inflammatoires/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Mémoire spatiale/effets des médicaments et des substances chimiques , Acetylcholinesterase/sang , Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/physiopathologie , Animaux , Encéphale/enzymologie , Encéphale/physiopathologie , Modèles animaux de maladie humaine , Femelle , Protéines liées au GPI/antagonistes et inhibiteurs , Protéines liées au GPI/sang , Médiateurs de l'inflammation/métabolisme , Mâle , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Souris transgéniques , Simulation de docking moléculaire , Maladies neuro-inflammatoires/enzymologie , Maladies neuro-inflammatoires/génétique , Maladies neuro-inflammatoires/physiopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Transduction du signal
20.
Life Sci ; 285: 119964, 2021 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-34537230

RÉSUMÉ

AIMS: Alzheimer's disease (AD) is the most common progressive neurodegenerative disorder characterized by declined cognitive functions in the elderly. Quercetin (Q) is a potent flavonol that has neuroprotective effects on AD derangements. The present study aimed to evaluate the α-secretase stimulatory function of Q through activation of ADAM10 and ADAM17 gene expression in the aluminum chloride (AlCl3)-induced AD rat model. MAIN METHODS: After induction of AD in rats by oral administration of AlCl3 (50 mg/kg) for 28 days, the Q doses (25 and 50 mg/kg) were orally administered for 28 days. Rats performed the behavioral assessments during the last week of the treatment period. Hippocampi were harvested for assessments of the neurochemical and histopathological examinations and gene expression analysis. KEY FINDINGS: Administration of Q to AlCl3-induced AD rat model attenuated behavioral deficits, improved cholinergic and dopaminergic dysfunctions, and diminished insoluble amyloid ß (Aß) plaques aggregation in the hippocampus. These ameliorative effects of Q were associated with down-regulation of APP, BACE1, APH1, and PSEN1 and up-regulation of ADAM10 and ADAM17 gene expression levels in the hippocampus. SIGNIFICANCE: The present study suggests that Q might attenuate neurotransmission impairment, Aß aggregation in the hippocampus, and behavioral deficits in the AlCl3-induce AD rat model via up-regulating ADAM 10 and ADAM 17 (α-secretase) gene expression, leading to the inhibition of the amyloidogenic pathway. In support of the present finding, we suggest that ADAM10 and ADAM17 activation might be potential drug targets for AD to counteract the Aß aggregation and cognitive deterioration.


Sujet(s)
Protéine ADAM10/métabolisme , Protéine ADAM17/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Amyloid precursor protein secretases/métabolisme , Peptides bêta-amyloïdes/antagonistes et inhibiteurs , Antioxydants/pharmacologie , Neuroprotecteurs/pharmacologie , Quercétine/pharmacologie , Protéine ADAM10/génétique , Protéine ADAM17/génétique , Chlorure d'aluminium , Maladie d'Alzheimer/induit chimiquement , Maladie d'Alzheimer/enzymologie , Amyloid precursor protein secretases/génétique , Peptides bêta-amyloïdes/métabolisme , Animaux , Antioxydants/usage thérapeutique , Modèles animaux de maladie humaine , Expression des gènes/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/enzymologie , Mâle , Neuroprotecteurs/usage thérapeutique , Quercétine/usage thérapeutique , Rats , Rat Wistar
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...