Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Cell Death Discov ; 10(1): 178, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627359

ABSTRACT

Mitochondrial dysfunction represents one of the most common molecular hallmarks of both sporadic and familial forms of amyotrophic lateral sclerosis (ALS), a neurodegenerative disorder caused by the selective degeneration and death of motor neurons. The accumulation of misfolded proteins on and within mitochondria, as observed for SOD1 G93A mutant, correlates with a drastic reduction of mitochondrial respiration and the inhibition of metabolites exchanges, including ADP/ATP and NAD+/NADH, across the Voltage-Dependent Anion-selective Channel 1 (VDAC1), the most abundant channel protein of the outer mitochondrial membrane. Here, we show that the AAV-mediated upregulation of VDAC1 in the spinal cord of transgenic mice expressing SOD1 G93A completely rescues the mitochondrial respiratory profile. This correlates with the increased activity and levels of key regulators of mitochondrial functions and maintenance, namely the respiratory chain Complex I and the sirtuins (Sirt), especially Sirt3. Furthermore, the selective increase of these mitochondrial proteins is associated with an increase in Tom20 levels, the receptor subunit of the TOM complex. Overall, our results indicate that the overexpression of VDAC1 has beneficial effects on ALS-affected tissue by stabilizing the Complex I-Sirt3 axis.

2.
Cells ; 13(8)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38667333

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by progressive cognitive decline and memory loss, imposing a significant burden on affected individuals and their families. Despite the recent promising progress in therapeutic approaches, more needs to be done to understand the intricate molecular mechanisms underlying the development and progression of AD. Growing evidence points to epigenetic changes as playing a pivotal role in the pathogenesis of the disease. The dynamic interplay between genetic and environmental factors influences the epigenetic landscape in AD, altering gene expression patterns associated with key pathological events associated with disease pathogenesis. To this end, epigenetic alterations not only impact the expression of genes implicated in AD pathogenesis but also contribute to the dysregulation of crucial cellular processes, including synaptic plasticity, neuroinflammation, and oxidative stress. Understanding the complex epigenetic mechanisms in AD provides new avenues for therapeutic interventions. This review comprehensively examines the role of DNA methylation and histone modifications in the context of AD. It aims to contribute to a deeper understanding of AD pathogenesis and facilitate the development of targeted therapeutic strategies.


Subject(s)
Alzheimer Disease , DNA Methylation , Epigenesis, Genetic , Histone Code , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , DNA Methylation/genetics , Histone Code/genetics , Histones/metabolism , Animals
3.
Microorganisms ; 12(4)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38674584

ABSTRACT

Biofilm (BF) can give rise to systemic infections, prolonged hospitalization times, and, in the worst case, death. This review aims to provide an overview of recent strategies for the prevention and destruction of pathogenic BFs. First, the main phases of the life cycle of BF and maturation will be described to identify potential targets for anti-BF approaches. Then, an approach acting on bacterial adhesion, quorum sensing (QS), and the extracellular polymeric substance (EPS) matrix will be introduced and discussed. Finally, bacteriophage-mediated strategies will be presented as innovative approaches against BF inhibition/destruction.

4.
Sci Rep ; 13(1): 18927, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37919374

ABSTRACT

Phage display is a molecular biology technique that allows the presentation of foreign peptides on the surface of bacteriophages. It is widely utilized for applications such as the discovery of biomarkers, the development of therapeutic antibodies, and the investigation of protein-protein interactions. When employing phages in diagnostic and therapeutic monitoring assays, it is essential to couple them with a detection system capable of revealing and quantifying the interaction between the peptide displayed on the phage capsid and the target of interest. This process is often technically challenging and costly. Here, we generated a fluorescent helper phage vector displaying sfGFP in-frame to the pIII of the capsid proteins. Further, we developed an exchangeable dual-display phage system by combining our newly developed fluorescent helper phage vector with a phagemid vector harboring the engineered pVIII with a peptide-probe. By doing so, the sfGFP and a peptide-probe are displayed on the same phage particle. Notably, our dual-display approach is highly flexible as it allows for easy exchange of the displayed peptide-probe on the pVIII to gain the desired selectivity, while maintaining the sfGFP gene, which allows easy visualization and quantification of the interaction peptide-probe. We anticipate that this system will reduce time and costs compared to the current phage-based detection systems.


Subject(s)
Bacteriophages , Bacteriophages/genetics , Bacteriophages/metabolism , Peptide Library , Peptides/chemistry , Capsid Proteins/genetics , Capsid/metabolism
5.
Biology (Basel) ; 12(9)2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37759663

ABSTRACT

Noradrenaline (NA) depletion occurs in Alzheimer's disease (AD); however, its relationship with the pathological expression of Tau and transactive response DNA-binding protein 43 (TDP-43), two major hallmarks of AD, remains elusive. Here, increasing doses of a selective noradrenergic immunotoxin were injected into developing rats to generate a model of mild or severe NA loss. At about 12 weeks post-lesion, dose-dependent working memory deficits were detected in these animals, associated with a marked increase in cortical and hippocampal levels of TDP-43 phosphorylated at Ser 409/410 and Tau phosphorylated at Thr 217. Notably, the total levels of both proteins were largely unaffected, suggesting a direct relationship between neocortical/hippocampal NA depletion and the phosphorylation of pathological Tau and TDP-43 proteins. As pTD43 is present in 23% of AD cases and pTau Thr217 has been detected in patients with mild cognitive impairment that eventually would develop into AD, improvement of noradrenergic function in AD might represent a viable therapeutic approach with disease-modifying potential.

6.
Biology (Basel) ; 12(3)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36979116

ABSTRACT

Large bone defect treatments have always been one of the important challenges in clinical practice and created a huge demand for more efficacious regenerative approaches. The bone tissue engineering (BTE) approach offered a new alternative to conventional bone grafts, addressing all clinical needs. Over the past years, BTE research is focused on the study and realisation of new biomaterials, including 3D-printed supports to improve mechanical, structural and biological properties. Among these, polylactic acid (PLA) scaffolds have been considered the most promising biomaterials due to their good biocompatibility, non-toxic biodegradability and bioresorbability. In this work, we evaluated the physiological response of human foetal osteoblast cells (hFOB), in terms of cell proliferation and osteogenic differentiation, within oxygen plasma treated 3D-printed PLA scaffolds, obtained by fused deposition modelling (FDM). A mechanical simulation to predict their behaviour to traction, flexural or torque solicitations was performed. We found that: 1. hFOB cells adhere and grow on scaffold surfaces; 2. hFOB grown on oxygen plasma treated PLA scaffolds (PLA_PT) show an improvement of cell adhesion and proliferation, compared to not-plasma treated scaffolds (PLA_NT); 3. Over time, hFOB penetrate along strands, differentiate, and form a fibrous matrix, tissue-like; 4. 3D-printed PLA scaffolds have good mechanical behaviour in each analysed configuration. These findings suggest that 3D-printed PLA scaffolds could represent promising biomaterials for medical implantable devices in the orthopaedic field.

7.
Biomolecules ; 13(3)2023 03 17.
Article in English | MEDLINE | ID: mdl-36979483

ABSTRACT

Alzheimer's disease (AD) is a common neurodegenerative disorder that affects the elderly. One of the key features of AD is the accumulation of reactive oxygen species (ROS), which leads to an overall increase in oxidative damage. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a master regulator of the antioxidant response in cells. Under low ROS levels, Nrf2 is kept in the cytoplasm. However, an increase in ROS production leads to a translocation of Nrf2 into the nucleus, where it activates the transcription of several genes involved in the cells' antioxidant response. Additionally, Nrf2 activation increases autophagy function. However, in AD, the accumulation of Aß and tau reduces Nrf2 levels, decreasing the antioxidant response. The reduced Nrf2 levels contribute to the further accumulation of Aß and tau by impairing their autophagy-mediated turnover. In this review, we discuss the overwhelming evidence indicating that genetic or pharmacological activation of Nrf2 is as a potential approach to mitigate AD pathology.


Subject(s)
Alzheimer Disease , Humans , Aged , Alzheimer Disease/drug therapy , NF-E2-Related Factor 2/metabolism , Antioxidants/therapeutic use , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Reactive Oxygen Species , Oxidative Stress
8.
Cell Death Dis ; 14(2): 122, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36792609

ABSTRACT

Mitochondrial dysfunction and the loss of mitophagy, aimed at recycling irreversibly damaged organelles, contribute to the onset of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease affecting spinal cord motor neurons. In this work, we showed that the reduction of mitochondrial respiration, exactly oxygen flows linked to ATP production and maximal capacity, correlates with the appearance of the most common ALS motor symptoms in a transgenic mouse model expressing SOD1 G93A mutant. This is the result of the equal inhibition in the respiration linked to complex I and II of the electron transport chain, but not their protein levels. Since the overall mitochondrial mass was unvaried, we investigated the expression of the Translocator Protein (TSPO), a small mitochondrial protein whose overexpression was recently linked to the loss of mitophagy in a model of Parkinson's disease. Here we clearly showed that levels of TSPO are significantly increased in ALS mice. Mechanistically, this increase is linked to the overactivation of ERK1/2 pathway and correlates with a decrease in the expression of the mitophagy-related marker Atg12, indicating the occurrence of impairments in the activation of mitophagy. Overall, our work sets out TSPO as a key regulator of mitochondrial homeostasis in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Animals , Mice , Amyotrophic Lateral Sclerosis/metabolism , Disease Models, Animal , MAP Kinase Signaling System , Mice, Transgenic , Mitochondria/metabolism , Mitophagy , Neurodegenerative Diseases/metabolism , Spinal Cord/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
9.
Int J Mol Sci ; 23(15)2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35955847

ABSTRACT

Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease represent some of the most prevalent neurodegenerative disorders afflicting millions of people worldwide. Unfortunately, there is a lack of efficacious treatments to cure or stop the progression of these disorders. While the causes of such a lack of therapies can be attributed to various reasons, the disappointing results of recent clinical trials suggest the need for novel and innovative approaches. Since its discovery, there has been a growing excitement around the potential for CRISPR-Cas9 mediated gene editing to identify novel mechanistic insights into disease pathogenesis and to mediate accurate gene therapy. To this end, the literature is rich with experiments aimed at generating novel models of these disorders and offering proof-of-concept studies in preclinical animal models validating the great potential and versatility of this gene-editing system. In this review, we provide an overview of how the CRISPR-Cas9 systems have been used in these neurodegenerative disorders.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Animals , CRISPR-Cas Systems/genetics , Gene Editing/methods , Genetic Therapy/methods , Humans , Neurodegenerative Diseases/drug therapy
10.
Biomedicines ; 9(8)2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34440152

ABSTRACT

Mutations in Cu/Zn Superoxide Dismutase (SOD1) gene represent one of the most common causes of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder that specifically affects motor neurons (MNs). The dismutase-active SOD1 G93A mutant is responsible for the formation of toxic aggregates onto the mitochondrial surface, using the Voltage-Dependent Anion Channel 1 (VDAC1) as an anchor point to the organelle. VDAC1 is the master regulator of cellular bioenergetics and by binding to hexokinases (HKs) it controls apoptosis. In ALS, however, SOD1 G93A impairs VDAC1 activity and displaces HK1 from mitochondria, promoting organelle dysfunction, and cell death. Using an ALS cell model, we demonstrate that a small synthetic peptide derived from the HK1 sequence (NHK1) recovers the cell viability in a dose-response manner and the defective mitochondrial respiration profile relative to the ADP phosphorylation. This correlates with an unexpected increase of VDAC1 expression and a reduction of SOD1 mutant accumulation at the mitochondrial level. Overall, our findings provide important new insights into the development of therapeutic molecules to fight ALS and help to better define the link between altered mitochondrial metabolism and MNs death in the disease.

11.
Mol Psychiatry ; 25(10): 2620-2629, 2020 10.
Article in English | MEDLINE | ID: mdl-30622336

ABSTRACT

The lack of effective treatments for Alzheimer's disease (AD) is alarming, considering the number of people currently affected by this disorder and the projected increase over the next few decades. Elevated homocysteine (Hcy) levels double the risk of developing AD. Choline, a primary dietary source of methyl groups, converts Hcy to methionine and reduces age-dependent cognitive decline. Here, we tested the transgenerational benefits of maternal choline supplementation (ChS; 5.0 g/kg choline chloride) in two generations (Gen) of APP/PS1 mice. We first exposed 2.5-month-old mice to the ChS diet and allowed them to breed with each other to generate Gen-1 mice. Gen-1 mice were exposed to the ChS diet only during gestation and lactation; once weaned at postnatal day 21, Gen-1 mice were then kept on the control diet for the remainder of their life. We also bred a subset of Gen-1 mice to each other and obtained Gen-2 mice; these mice were never exposed to ChS. We found that ChS reduced Aß load and microglia activation, and improved cognitive deficits in old Gen-1 and Gen-2 APP/PS1 mice. Mechanistically, these changes were linked to a reduction in brain Hcy levels in both generations. Further, RNA-Seq data from APP/PS1 hippocampal tissue revealed that ChS significantly changed the expression of 27 genes. These genes were enriched for inflammation, histone modifications, and neuronal death functional classes. Our results are the first to demonstrate a transgenerational benefit of ChS and suggest that modifying the maternal diet with additional choline reduces AD pathology across multiple generations.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Brain/metabolism , Choline/pharmacology , Dietary Supplements , Homocysteine/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Choline/administration & dosage , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic
12.
Aging Cell ; 18(1): e12873, 2019 02.
Article in English | MEDLINE | ID: mdl-30488653

ABSTRACT

Accumulation of amyloid-ß (Aß) and fibrillary tangles, as well as neuroinflammation and memory loss, are hallmarks of Alzheimer's disease (AD). After almost 15 years from their generation, 3xTg-AD mice are still one of the most used transgenic models of AD. Converging evidence indicates that the phenotype of 3xTg-AD mice has shifted over the years and contradicting reports about onset of pathology or cognitive deficits are apparent in the literature. Here, we assessed Aß and tau load, neuroinflammation, and cognitive changes in 2-, 6-, 12-, and 20-month-old female 3xTg-AD and nontransgenic (NonTg) mice. We found that ~80% of the mice analyzed had Aß plaques in the caudal hippocampus at 6 months of age, while 100% of them had Aß plaques in the hippocampus at 12 months of age. Cortical Aß plaques were first detected at 12 months of age, including in the entorhinal cortex. Phosphorylated Tau at Ser202/Thr205 and Ser422 was apparent in the hippocampus of 100% of 6-month-old mice, while only 50% of mice showed tau phosphorylation at Thr212/Ser214 at this age. Neuroinflammation was first evident in 6-month-old mice and increased as a function of age. These neuropathological changes were clearly associated with progressive cognitive decline, which was first apparent at 6 months of age and became significantly worse as the mice aged. These data indicate a consistent and predictable progression of the AD-like pathology in female 3xTg-AD mice, and will facilitate the design of future studies using these mice.


Subject(s)
Alzheimer Disease/pathology , Disease Progression , Aging/pathology , Alzheimer Disease/complications , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Gliosis/pathology , Hippocampus/metabolism , Hippocampus/pathology , Memory Disorders/complications , Memory Disorders/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Phosphorylation , Plaque, Amyloid/pathology , Time Factors , tau Proteins/metabolism
13.
Neurobiol Aging ; 68: 1, 2018 08.
Article in English | MEDLINE | ID: mdl-29729422

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease. The causes of sporadic AD, which represents more than 95% of AD cases, are unknown. Several AD risk factors have been identified and among these, type 2 diabetes increases the risk of developing AD by 2-fold. However, the mechanisms by which diabetes contributes to AD pathogenesis remain elusive. The mammalian target of rapamycin (mTOR) is a protein kinase that plays a crucial role in the insulin signaling pathway and has been linked to AD. We used a crossbreeding strategy to remove 1 copy of the mTOR gene from the forebrain of Tg2576 mice, a mouse model of AD. We used 20-month-old mice to assess changes in central insulin signaling and found that Tg2576 mice had impaired insulin signaling. These impairments were mTOR dependent as we found an improvement in central insulin signaling in mice with lower brain mTOR activity. Furthermore, removing 1 copy of mTOR from Tg2576 mice improved cognition and reduced levels of Aß, tau, and cytokines. Our findings indicate that mTOR signaling is a key mediator of central insulin dysfunction in Tg2576. These data further highlight a possible role for mTOR signaling in AD pathogenesis and add to the body of evidence indicating that reducing mTOR activity could be a valid therapeutic approach for AD.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/genetics , Gene Dosage , Insulin/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cognition , Cytokines/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Mice, Transgenic , Molecular Targeted Therapy , TOR Serine-Threonine Kinases/physiology , tau Proteins/metabolism
14.
Hum Mol Genet ; 26(24): 4823-4835, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29036636

ABSTRACT

Aging is the major risk factor for several neurodegenerative diseases, including Alzheimer's disease (AD). However, the mechanisms by which aging contributes to neurodegeneration remain elusive. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a transcription factor that regulates expression of a vast number of genes by binding to the antioxidant response element. Nrf2 levels decrease as a function of age, and reduced Nrf2 levels have been reported in postmortem human brains and animal models of AD. Nevertheless, it is still unknown whether Nrf2 plays a role in the cognitive deficits associated with AD. To address this question, we used a genetic approach to remove the Nrf2 gene from APP/PS1 mice, a widely used animal model of AD. We found that the lack of Nrf2 significantly exacerbates cognitive deficits in APP/PS1, without altering gross motor function. Specifically, we found an exacerbation of deficits in spatial learning and memory, as well as in working and associative memory. Different brain regions control these behavioral tests, indicating that the lack of Nrf2 has a global effect on brain function. The changes in cognition were linked to an increase in Aß and interferon-gamma (IFNγ) levels, and microgliosis. The changes in IFNγ levels are noteworthy as previously published evidence indicates that IFNγ can increase microglia activation and induce Aß production. Our data suggest a clear link between Nrf2 and AD-mediated cognitive decline and further strengthen the connection between Nrf2 and AD.


Subject(s)
Alzheimer Disease/genetics , Cognition Disorders/genetics , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Cognition Disorders/metabolism , Disease Models, Animal , Female , Male , Memory Disorders/genetics , Mice , Mice, Transgenic , NF-E2-Related Factor 2/metabolism , Presenilin-1/genetics
15.
Aging Cell ; 16(5): 1146-1154, 2017 10.
Article in English | MEDLINE | ID: mdl-28779511

ABSTRACT

There is an urgent need for the development of new therapeutic strategies for Alzheimer's disease (AD). The dual-specificity tyrosine phosphorylation-regulated kinase-1A (Dyrk1a) is a protein kinase that phosphorylates the amyloid precursor protein (APP) and tau and thus represents a link between two key proteins involved in AD pathogenesis. Furthermore, Dyrk1a is upregulated in postmortem human brains, and high levels of Dyrk1a are associated with mental retardation. Here, we sought to determine the effects of Dyrk1 inhibition on AD-like pathology developed by 3xTg-AD mice, a widely used animal model of AD. We dosed 10-month-old 3xTg-AD and nontransgenic (NonTg) mice with a Dyrk1 inhibitor (Dyrk1-inh) or vehicle for eight weeks. During the last three weeks of treatment, we tested the mice in a battery of behavioral tests. The brains were then analyzed for the pathological markers of AD. We found that chronic Dyrk1 inhibition reversed cognitive deficits in 3xTg-AD mice. These effects were associated with a reduction in amyloid-ß (Aß) and tau pathology. Mechanistically, Dyrk1 inhibition reduced APP and insoluble tau phosphorylation. The reduction in APP phosphorylation increased its turnover and decreased Aß levels. These results suggest that targeting Dyrk1 could represent a new viable therapeutic approach for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/genetics , Cognitive Dysfunction/prevention & control , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , tau Proteins/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Animals , Benzimidazoles/pharmacology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Locomotion/drug effects , Maze Learning/drug effects , Mice , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Signal Transduction , tau Proteins/antagonists & inhibitors , tau Proteins/metabolism , Dyrk Kinases
16.
Nat Neurosci ; 20(9): 1236-1246, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28758999

ABSTRACT

Alzheimer's disease (AD) is characterized by severe neuronal loss; however, the mechanisms by which neurons die remain elusive. Necroptosis, a programmed form of necrosis, is executed by the mixed lineage kinase domain-like (MLKL) protein, which is triggered by receptor-interactive protein kinases (RIPK) 1 and 3. We found that necroptosis was activated in postmortem human AD brains, positively correlated with Braak stage, and inversely correlated with brain weight and cognitive scores. In addition, we found that the set of genes regulated by RIPK1 overlapped significantly with multiple independent AD transcriptomic signatures, indicating that RIPK1 activity could explain a substantial portion of transcriptomic changes in AD. Furthermore, we observed that lowering necroptosis activation reduced cell loss in a mouse model of AD. We anticipate that our findings will spur a new area of research in the AD field focused on developing new therapeutic strategies aimed at blocking its activation.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Apoptosis/physiology , Temporal Lobe/metabolism , Temporal Lobe/pathology , Alzheimer Disease/genetics , Animals , Cells, Cultured , Humans , Maze Learning/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Necrosis/metabolism , Necrosis/pathology , Random Allocation
17.
Mol Neurodegener ; 11(1): 52, 2016 07 13.
Article in English | MEDLINE | ID: mdl-27412291

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder worldwide. Clinically, AD is characterized by impairments of memory and cognitive functions. Accumulation of amyloid-ß (Aß) and neurofibrillary tangles are the prominent neuropathologies in patients with AD. Strong evidence indicates that an imbalance between production and degradation of key proteins contributes to the pathogenesis of AD. The mammalian target of rapamycin (mTOR) plays a key role in maintaining protein homeostasis as it regulates both protein synthesis and degradation. A key regulator of mTOR activity is the proline-rich AKT substrate 40 kDa (PRAS40), which directly binds to mTOR and reduces its activity. Notably, AD patients have elevated levels of phosphorylated PRAS40, which correlate with Aß and tau pathologies as well as cognitive deficits. Physiologically, PRAS40 phosphorylation is regulated by Pim1, a protein kinase of the protoconcogene family. Here, we tested the effects of a selective Pim1 inhibitor (Pim1i), on spatial reference and working memory and AD-like pathology in 3xTg-AD mice. RESULTS: We have identified a Pim1i that crosses the blood brain barrier and reduces PRAS40 phosphorylation. Pim1i-treated 3xTg-AD mice performed significantly better than their vehicle treated counterparts as well as non-transgenic mice. Additionally, 3xTg-AD Pim1i-treated mice showed a reduction in soluble and insoluble Aß40 and Aß42 levels, as well as a 45.2 % reduction in Aß42 plaques within the hippocampus. Furthermore, phosphorylated tau immunoreactivity was reduced in the hippocampus of Pim1i-treated 3xTg-AD mice by 38 %. Mechanistically, these changes were linked to a significant increase in proteasome activity. CONCLUSION: These results suggest that reductions in phosphorylated PRAS40 levels via Pim1 inhibition reduce Aß and Tau pathology and rescue cognitive deficits by increasing proteasome function. Given that Pim1 inhibitors are already being tested in ongoing human clinical trials for cancer, the results presented here may open a new venue of drug discovery for AD by developing more Pim1 inhibitors.


Subject(s)
Alzheimer Disease/metabolism , Cognition/physiology , Hippocampus/metabolism , Neurofibrillary Tangles/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/metabolism , Animals , Cognition Disorders/metabolism , Disease Models, Animal , Humans , Memory/physiology , Mice , Proto-Oncogene Proteins c-pim-1/metabolism
18.
J Neurosci ; 35(41): 14042-56, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468204

ABSTRACT

Aging is the most important risk factor associated with Alzheimer's disease (AD); however, the molecular mechanisms linking aging to AD remain unclear. Suppression of the ribosomal protein S6 kinase 1 (S6K1) increases healthspan and lifespan in several organisms, from nematodes to mammals. Here we show that S6K1 expression is upregulated in the brains of AD patients. Using a mouse model of AD, we found that genetic reduction of S6K1 improved synaptic plasticity and spatial memory deficits, and reduced the accumulation of amyloid-ß and tau, the two neuropathological hallmarks of AD. Mechanistically, these changes were linked to reduced translation of tau and the ß-site amyloid precursor protein cleaving enzyme 1, a key enzyme in the generation of amyloid-ß. Our results implicate S6K1 dysregulation as a previously unidentified molecular mechanism underlying synaptic and memory deficits in AD. These findings further suggest that therapeutic manipulation of S6K1 could be a valid approach to mitigate AD pathology. SIGNIFICANCE STATEMENT: Aging is the most important risk factor for Alzheimer's disease (AD). However, little is known about how it contributes to AD pathogenesis. S6 kinase 1 (S6K1) is a protein kinase involved in regulation of protein translation. Reducing S6K1 activity increases lifespan and healthspan. We report the novel finding that reducing S6K1 activity in 3xTg-AD mice ameliorates synaptic and cognitive deficits. These improvement were associated with a reduction in amyloid-ß and tau pathology. Mechanistically, lowering S6K1 levels reduced translation of ß-site amyloid precursor protein cleaving enzyme 1 and tau, two key proteins involved in AD pathogenesis. These data suggest that S6K1 may represent a molecular link between aging and AD. Given that aging is the most important risk factor for most neurodegenerative diseases, our results may have far-reaching implications into other diseases.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Gene Expression Regulation/physiology , Memory Disorders/therapy , Neuronal Plasticity/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Hippocampus/pathology , Humans , Locomotion/genetics , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Maze Learning/physiology , Memory Disorders/etiology , Mice , Mice, Transgenic , Neuronal Plasticity/genetics , Neurons/physiology , Peptide Fragments/metabolism , Presenilin-1/metabolism , Proteasome Endopeptidase Complex/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction/genetics , tau Proteins/genetics , tau Proteins/metabolism
19.
Hum Mol Genet ; 24(16): 4625-35, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26002100

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD-TDP) are two neurodegenerative disorders characterized by the accumulation of TDP-43. TDP-43 is proteolitically cleaved to generate two major C-terminal fragments of 35 and 25 kDa. The latter, known as TDP-25, is a consistent feature of FTLD-TDP and ALS; however, little is known about its role in disease pathogenesis. We have previously developed transgenic mice overexpressing low levels of TDP-25 (TgTDP-25(+/0)), which at 6 months of age show mild cognitive impairments and no motor deficits. To better understand the role of TDP-25 in the pathogenesis of ALS and FTLD-TDP, we generated TDP-25 homozygous mice (TgTDP-25(+/+)), thereby further increasing TDP-25 expression. We found a gene-dosage effect on cognitive and motor function at 15 months of age, as the TgTDP-25(+/+) showed more severe spatial and working memory deficits as well as worse motor performance than TgTDP-25(+/0) mice. These behavioral deficits were associated with increased soluble levels of TDP-25 in the nucleus and cytosol. Notably, high TDP-25 levels were also linked to reduced autophagy induction and proteasome function, two events that have been associated with both ALS and FTLD-TDP. In summary, we present strong in vivo evidence that high levels of TDP-25 are sufficient to cause behavioral deficits and reduce function of two of the major protein turnover systems: autophagy and proteasome. These mice represent a new tool to study the role of TDP-25 in the pathogenesis of ALS and FTLD-TDP.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/metabolism , Proteolysis , Amyotrophic Lateral Sclerosis/genetics , Animals , Autophagy/genetics , Behavior, Animal , DNA-Binding Proteins/genetics , Frontotemporal Lobar Degeneration/genetics , Humans , Mice , Mice, Transgenic , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary
20.
Neurobiol Aging ; 35(12): 2726-2735, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25034342

ABSTRACT

Currently, there are no available approaches to cure or slow down the progression of Alzheimer's disease (AD), which is characterized by the accumulation of extracellular amyloid-ß (Aß) deposits and intraneuronal tangles that comprised hyperphosphorylated tau. The ß2 adrenergic receptors (ß2ARs) are expressed throughout the cortex and hippocampus and play a key role in cognitive functions. Alterations in the function of these receptors have been linked to AD; however, these data remain controversial as apparent contradicting reports have been published. Given the current demographics of growing elderly population and the high likelihood of concurrent ß-blocker use for other chronic conditions, more studies into the role of this receptor in AD animal models are needed. Here, we show that administration of ICI 118,551 (ICI), a selective ß2AR antagonist, exacerbates cognitive deficits in a mouse model of AD, the 3xTg-AD mice. Neuropathologically, ICI increased Aß levels and Aß plaque burden. Concomitantly, ICI-treated 3xTg-AD mice showed an increase in tau phosphorylation and accumulation. Mechanistically, these changes were linked to an increase in amyloidogenic amyloid precursor protein processing. These results suggest that under the conditions used here, selective pharmacologic inhibition of ß2ARs has detrimental effects on AD-like pathology in mice. Overall, these studies strengthen the notion that the link between ß2ARs and AD is likely highly complex and suggest caution in generalizing the beneficial effects of ß blockers on AD.


Subject(s)
Adrenergic beta-2 Receptor Antagonists/adverse effects , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Cognition , Propanolamines/adverse effects , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Antagonists/administration & dosage , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Disease Progression , Female , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice, Transgenic , Phosphorylation/drug effects , Propanolamines/administration & dosage , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...