Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Alzheimers Res Ther ; 16(1): 141, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943196

ABSTRACT

BACKGROUND: Amyloid-ß (Aß) and tau are brain hallmarks of Alzheimer's disease (AD), also present in blood as soluble biomarkers or encapsulated in extracellular vesicles (EVs). Our goal was to assess how soluble plasma biomarkers of AD pathology correlate with the number and content of EVs. METHODS: Single-molecule enzyme-linked assays were used to quantify Aß42/40 and tau in plasma samples and neurally-derived EVs (NDEVs) from a cohort of APOE ε4- (n = 168) and APOE ε4+ (n = 68) cognitively normal individuals and AD patients (n = 55). The ratio of CD56 (Neuronal cell-adhesion molecule) to CD81 signal measured by ELISA-DELFIA was used for the relative quantification of NDEVs in plasma samples. RESULTS: The soluble plasma Aß42/40 ratio is decreased in AD patients compared to cognitively normal individuals. The amount and content (Aß40, Aß42, tau) of plasma NDEVs were similar between groups. Plasma NDEVs quantity remain consistent with aging and between AD and CN individuals. However, the quantity of soluble biomarkers was negatively correlated to NDEVs number in cognitively normal individuals, while in AD patients, this correlation is lost, suggesting a shift in the mechanism underpinning the production and the release of these biomarkers in pathological conditions. CONCLUSION: Soluble plasma Aß42/40 ratio is the most robust biomarker to discriminate between AD patients and CN individuals, as it normalizes for the number of NDEVs. Analysis of NDEVs and their content pointed toward peculiar mechanisms of Aß release in AD. Further research on independent cohorts can confirm our findings and assess whether plasma Aß and tau need correction by NDEVs for better AD risk identification in CN populations.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Biomarkers , Extracellular Vesicles , tau Proteins , Humans , Alzheimer Disease/blood , Extracellular Vesicles/metabolism , Biomarkers/blood , Female , Male , Amyloid beta-Peptides/blood , Aged , tau Proteins/blood , Peptide Fragments/blood , Aged, 80 and over , Middle Aged , Cohort Studies , Apolipoprotein E4/genetics , Apolipoprotein E4/blood
2.
Int J Mol Sci ; 25(2)2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38256246

ABSTRACT

Alzheimer's disease (AD) is characterized by amyloid beta (Aß) plaques and hyperphosphorylated tau in the brain. Aß plaques precede cognitive impairments and can be detected through amyloid-positron emission tomography (PET) or in cerebrospinal fluid (CSF). Assessing the plasma Aß42/Aß40 ratio seems promising for non-invasive and cost-effective detection of brain Aß accumulation. This approach involves some challenges, including the accuracy of blood-based biomarker measurements and the establishment of clear, standardized thresholds to categorize the risk of developing brain amyloid pathology. Plasma Aß42/Aß40 ratio was measured in 277 volunteers without dementia, 70 AD patients and 18 non-AD patients using single-molecule array. Patients (n = 88) and some volunteers (n = 66) were subject to evaluation of amyloid status by CSF Aß quantification or PET analysis. Thresholds of plasma Aß42/Aß40 ratio were determined based on a Gaussian mixture model, a decision tree, and the Youden's index. The 0.0472 threshold, the one with the highest sensitivity, was retained for general population without dementia screening, and the 0.0450 threshold was retained for research and clinical trials recruitment, aiming to minimize the need for CSF or PET analyses to identify amyloid-positive individuals. These findings offer a promising step towards a cost-effective method for identifying individuals at risk of developing AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Humans , Amyloidogenic Proteins , Positron-Emission Tomography , Alzheimer Disease/diagnosis , Brain , Plaque, Amyloid
3.
Nat Commun ; 14(1): 3706, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37349319

ABSTRACT

Tau protein aggregates in several neurodegenerative disorders, referred to as tauopathies. The tau isoforms observed in post mortem human brain aggregates is used to classify tauopathies. However, distinguishing tauopathies ante mortem remains challenging, potentially due to differences between insoluble tau in aggregates and soluble tau in body fluids. Here, we demonstrated that tau isoforms differ between tauopathies in insoluble aggregates, but not in soluble brain extracts. We therefore characterized post-translational modifications of both the aggregated and the soluble tau protein obtained from post mortem human brain tissue of patients with Alzheimer's disease, cortico-basal degeneration, Pick's disease, and frontotemporal lobe degeneration. We found specific soluble signatures for each tauopathy and its specific aggregated tau isoforms: including ubiquitination on Lysine 369 for cortico-basal degeneration and acetylation on Lysine 311 for Pick's disease. These findings provide potential targets for future development of fluid-based biomarker assays able to distinguish tauopathies in vivo.


Subject(s)
Alzheimer Disease , Corticobasal Degeneration , Pick Disease of the Brain , Tauopathies , Humans , tau Proteins/metabolism , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Pick Disease of the Brain/metabolism , Lysine/metabolism , Tauopathies/diagnosis , Tauopathies/metabolism , Protein Isoforms/metabolism , Brain/metabolism , Protein Processing, Post-Translational
4.
Acta Neuropathol Commun ; 11(1): 82, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37198698

ABSTRACT

Aging is the main risk factor for Alzheimer's disease (AD) and other neurodegenerative pathologies, but the molecular and cellular changes underlying pathological aging of the nervous system are poorly understood. AD pathology seems to correlate with the appearance of cells that become senescent due to the progressive accumulation of cellular insults causing DNA damage. Senescence has also been shown to reduce the autophagic flux, a mechanism involved in clearing damaged proteins from the cell, and such impairment has been linked to AD pathogenesis. In this study, we investigated the role of cellular senescence on AD pathology by crossing a mouse model of AD-like amyloid-ß (Aß) pathology (5xFAD) with a mouse model of senescence that is genetically deficient for the RNA component of the telomerase (Terc-/-). We studied changes in amyloid pathology, neurodegeneration, and the autophagy process in brain tissue samples and primary cultures derived from these mice by complementary biochemical and immunostaining approaches. Postmortem human brain samples were also processed to evaluate autophagy defects in AD patients. Our results show that accelerated senescence produces an early accumulation of intraneuronal Aß in the subiculum and cortical layer V of 5xFAD mice. This correlates with a reduction in amyloid plaques and Aß levels in connecting brain regions at a later disease stage. Neuronal loss was specifically observed in brain regions presenting intraneuronal Aß and was linked to telomere attrition. Our results indicate that senescence affects intraneuronal Aß accumulation by impairing autophagy function and that early autophagy defects can be found in the brains of AD patients. Together, these findings demonstrate the instrumental role of senescence in intraneuronal Aß accumulation, which represents a key event in AD pathophysiology, and emphasize the correlation between the initial stages of amyloid pathology and defects in the autophagy flux.


Subject(s)
Alzheimer Disease , Neurons , Humans , Mice , Animals , Neurons/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/pathology , Brain/pathology , Autophagy , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic , Disease Models, Animal
5.
Biomedicines ; 10(11)2022 Oct 29.
Article in English | MEDLINE | ID: mdl-36359274

ABSTRACT

Most neurodegenerative diseases have the characteristics of protein folding disorders, i.e., they cause lesions to appear in vulnerable regions of the nervous system, corresponding to protein aggregates that progressively spread through the neuronal network as the symptoms progress. Alzheimer's disease is one of these diseases. It is characterized by two types of lesions: neurofibrillary tangles (NFTs) composed of tau proteins and senile plaques, formed essentially of amyloid peptides (Aß). A combination of factors ranging from genetic mutations to age-related changes in the cellular context converge in this disease to accelerate Aß deposition. Over the last two decades, numerous studies have attempted to elucidate how structural determinants of its precursor (APP) modify Aß production, and to understand the processes leading to the formation of different Aß aggregates, e.g., fibrils and oligomers. The synthesis proposed in this review indicates that the same motifs can control APP function and Aß production essentially by regulating membrane protein dimerization, and subsequently Aß aggregation processes. The distinct properties of these motifs and the cellular context regulate the APP conformation to trigger the transition to the amyloid pathology. This concept is critical to better decipher the patterns switching APP protein conformation from physiological to pathological and improve our understanding of the mechanisms underpinning the formation of amyloid fibrils that devastate neuronal functions.

6.
Mol Neurobiol ; 59(12): 7466-7485, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36197591

ABSTRACT

Neurotrypsin (NT) is a highly specific nervous system multi-domain serine protease best known for its selective processing of the potent synaptic organizer agrin. Its enzymatic activity is thought to influence processes of synaptic plasticity, with its deregulation causing accelerated neuromuscular junction (NMJ) degeneration or contributing to forms of mental retardation. These biological effects are likely to stem from NT-based regulation of agrin signaling. However, dissecting the exact biological implications of NT-agrin interplay is difficult, due to the scarce molecular detail regarding NT activity and NT-agrin interactions. We developed a strategy to reliably produce and purify a catalytically competent engineered variant of NT called "NT-mini" and a library of C-terminal agrin fragments, with which we performed a thorough biochemical and biophysical characterization of NT enzyme functionality. We studied the regulatory effects of calcium ions and heparin, identified NT's heparin-binding domain, and discovered how zinc ions induce modulation of enzymatic activity. Additionally, we investigated myotube differentiation and hippocampal neuron excitability, evidencing a dose-dependent increase in neuronal activity alongside a negative impact on myoblast fusion when using the active NT enzyme. Collectively, our results provide in vitro and cellular foundations to unravel the molecular underpinnings and biological significance of NT-agrin interactions.


Subject(s)
Agrin , Muscle Fibers, Skeletal , Agrin/chemistry , Neurons , Heparin , Synapses
7.
Mol Neurobiol ; 58(12): 6647-6669, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34608607

ABSTRACT

The ß-amyloid peptide (Aß) is found as amyloid fibrils in senile plaques, a typical hallmark of Alzheimer's disease (AD). However, intermediate soluble oligomers of Aß are now recognized as initiators of the pathogenic cascade leading to AD. Studies using recombinant Aß have shown that hexameric Aß in particular acts as a critical nucleus for Aß self-assembly. We recently isolated hexameric Aß assemblies from a cellular model, and demonstrated their ability to enhance Aß aggregation in vitro. Here, we report the presence of similar hexameric-like Aß assemblies across several cellular models, including neuronal-like cell lines. In order to better understand how they are produced in a cellular context, we investigated the role of presenilin-1 (PS1) and presenilin-2 (PS2) in their formation. PS1 and PS2 are the catalytic subunits of the γ-secretase complex that generates Aß. Using CRISPR-Cas9 to knockdown each of the two presenilins in neuronal-like cell lines, we observed a direct link between the PS2-dependent processing pathway and the release of hexameric-like Aß assemblies in extracellular vesicles. Further, we assessed the contribution of hexameric Aß to the development of amyloid pathology. We report the early presence of hexameric-like Aß assemblies in both transgenic mice brains exhibiting human Aß pathology and in the cerebrospinal fluid of AD patients, suggesting hexameric Aß as a potential early AD biomarker. Finally, cell-derived hexameric Aß was found to seed other human Aß forms, resulting in the aggravation of amyloid deposition in vivo and neuronal toxicity in vitro.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Plaque, Amyloid/metabolism , Presenilins/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , CHO Cells , Cell Line, Tumor , Cricetulus , Fibroblasts/metabolism , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/pathology
8.
Sci Rep ; 11(1): 17600, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34475508

ABSTRACT

The function of the amyloid precursor protein (APP) is not fully understood, but its cleavage product amyloid beta (Aß) together with neurofibrillary tangles constitute the hallmarks of Alzheimer's disease (AD). Yet, imbalance of excitatory and inhibitory neurotransmission accompanied by loss of synaptic functions, has been reported much earlier and independent of any detectable pathological markers. Recently, soluble APP fragments have been shown to bind to presynaptic GABAB receptors (GABABRs), subsequently decreasing the probability of neurotransmitter release. In this body of work, we were able to show that overexpression of wild-type human APP in mice (hAPPwt) causes early cognitive impairment, neuronal loss, and electrophysiological abnormalities in the absence of amyloid plaques and at very low levels of Aß. hAPPwt mice exhibited neuronal overexcitation that was evident in EEG and increased long-term potentiation (LTP). Overexpression of hAPPwt did not alter GABAergic/glutamatergic receptor components or GABA production ability. Nonetheless, we detected a decrease of GABA but not glutamate that could be linked to soluble APP fragments, acting on presynaptic GABABRs and subsequently reducing GABA release. By using a specific presynaptic GABABR antagonist, we were able to rescue hyperexcitation in hAPPwt animals. Our results provide evidence that APP plays a crucial role in regulating inhibitory neurotransmission.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Receptors, Glutamate/metabolism , Up-Regulation , gamma-Aminobutyric Acid/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Humans , Male , Mice , Neuronal Plasticity , Synapses/genetics , Synapses/metabolism , Synaptic Transmission
9.
Sci Rep ; 11(1): 11570, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34078941

ABSTRACT

A key hallmark of Alzheimer's disease is the extracellular deposition of amyloid plaques composed primarily of the amyloidogenic amyloid-ß (Aß) peptide. The Aß peptide is a product of sequential cleavage of the Amyloid Precursor Protein, the first step of which gives rise to a C-terminal Fragment (C99). Cleavage of C99 by γ-secretase activity releases Aß of several lengths and the Aß42 isoform in particular has been identified as being neurotoxic. The misfolding of Aß leads to subsequent amyloid fibril formation by nucleated polymerisation. This requires an initial and critical nucleus for self-assembly. Here, we identify and characterise the composition and self-assembly properties of cell-derived hexameric Aß42 and show its assembly enhancing properties which are dependent on the Aß monomer availability. Identification of nucleating assemblies that contribute to self-assembly in this way may serve as therapeutic targets to prevent the formation of toxic oligomers.


Subject(s)
Amyloid beta-Peptides/chemistry , Biopolymers/chemistry , Animals , CHO Cells , Cricetulus
11.
iScience ; 23(12): 101887, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33367225

ABSTRACT

Amyloid precursor protein (APP) cleavage by the ß-secretase produces the C99 transmembrane (TM) protein, which contains three dimerization-inducing Gly-x-x-x-Gly motifs. We demonstrate that dimeric C99 TM orientations regulate the precise cleavage lines by γ-secretase. Of all possible dimeric orientations imposed by a coiled-coil to the C99 TM domain, the dimer containing the 33Gly-x-x-x-Gly37 motif in the interface promoted the Aß42 processing line and APP intracellular domain-dependent gene transcription, including the induction of BACE1 mRNA, enhancing amyloidogenic processing and signaling. Another orientation exhibiting the 25Gly-x-x-x-Gly29 motif in the interface favored processing to Aß43/40. It induced significantly less gene transcription, while promoting formation of SDS-resistant "Aß-like" oligomers, reminiscent of Aß peptide oligomers. These required both Val24 of a pro-ß motif and the 25Gly-x-x-x-Gly29 interface. Thus, crossing angles imposed by precise dimeric orientations control γ-secretase initial cleavage at Aß48 or Aß49, linking the former to enhanced signaling and Aß42 production.

12.
Int J Mol Sci ; 22(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374485

ABSTRACT

The neuromuscular junction (NMJ) is at the crossroad between the nervous system (NS) and the muscle. Following neurotransmitter release from the motor neurons (MNs), muscle contraction occurs and movement is generated. Besides eliciting muscle contraction, the NMJ represents a site of chemical bidirectional interplay between nerve and muscle with the active participation of Schwann cells. Indeed, signals originating from the muscle play an important role in synapse formation, stabilization, maintenance and function, both in development and adulthood. We focus here on the contribution of the Glial cell line-Derived Neurotrophic Factor (GDNF) to these processes and to its potential role in the protection of the NMJ during neurodegeneration. Historically related to the maintenance and survival of dopaminergic neurons of the substantia nigra, GDNF also plays a fundamental role in the peripheral NS (PNS). At this level, it promotes muscle trophism and it participates to the functionality of synapses. Moreover, compared to the other neurotrophic factors, GDNF shows unique peculiarities, which make its contribution essential in neurodegenerative disorders. While describing the known structural and functional changes occurring at the NMJ during neurodegeneration, we highlight the role of GDNF in the NMJ-muscle cross-talk and we review its therapeutic potential in counteracting the degenerative process occurring in the PNS in progressive and severe diseases such as Alzheimer's disease (AD), Amyotrophic Lateral Sclerosis (ALS) and Spinal Muscular Atrophy (SMA). We also describe functional 3D neuromuscular co-culture systems that have been recently developed as a model for studying both NMJ formation in vitro and its involvement in neuromuscular disorders.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/physiology , Neuromuscular Junction/physiopathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Mice , Motor Neurons , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/physiopathology , Neurodegenerative Diseases/metabolism , Rats , Substantia Nigra/metabolism , Synapses/metabolism , Synaptic Transmission
13.
eNeuro ; 7(3)2020.
Article in English | MEDLINE | ID: mdl-32327470

ABSTRACT

The amyloid precursor protein (APP) has been extensively studied as the precursor of the ß-amyloid (Aß) peptide, the major component of the senile plaques found in the brain of Alzheimer's disease (AD) patients. However, the function of APP per se in neuronal physiology remains to be fully elucidated. APP is expressed at high levels in the brain. It resembles a cell adhesion molecule or a membrane receptor, suggesting that its function relies on cell-cell interaction and/or activation of intracellular signaling pathways. In this respect, the APP intracellular domain (AICD) was reported to act as a transcriptional regulator. Here, we used a transcriptome-based approach to identify the genes transcriptionally regulated by APP in the rodent embryonic cortex and on maturation of primary cortical neurons. Surprisingly, the overall transcriptional changes were subtle, but a more detailed analysis pointed to genes clustered in neuronal-activity dependent pathways. In particular, we observed a decreased transcription of neuronal PAS domain protein 4 (NPAS4) in APP-/- neurons. NPAS4 is an inducible transcription factor (ITF) regulated by neuronal depolarization. The downregulation of NPAS4 co-occurs with an increased production of the inhibitory neurotransmitter GABA and a reduced expression of the GABAA receptors α1. CRISPR-Cas-mediated silencing of NPAS4 in neurons led to similar observations. Patch-clamp investigation did not reveal any functional decrease of GABAA receptors activity, but long-term potentiation (LTP) measurement supported an increased GABA component in synaptic transmission of APP-/- mice. Together, NPAS4 appears to be a downstream target involved in APP-dependent regulation of inhibitory synaptic transmission.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Alzheimer Disease/genetics , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Humans , Mice , Synaptic Transmission , Transcription Factors , gamma-Aminobutyric Acid
14.
Front Neurosci ; 14: 586108, 2020.
Article in English | MEDLINE | ID: mdl-33551720

ABSTRACT

Presenilin 1 (PS1) and Presenilin 2 (PS2) are predominantly known as the catalytic subunits of the γ-secretase complex that generates the amyloid-ß (Aß) peptide, the major constituent of the senile plaques found in the brain of Alzheimer's disease (AD) patients. Apart from their role in γ-secretase activity, a growing number of cellular functions have been recently attributed to PSs. Notably, PSs were found to be enriched in mitochondria-associated membranes (MAMs) where mitochondria and endoplasmic reticulum (ER) interact. PS2 was more specifically reported to regulate calcium shuttling between these two organelles by controlling the formation of functional MAMs. We have previously demonstrated in mouse embryonic fibroblasts (MEF) an altered mitochondrial morphology along with reduced mitochondrial respiration and increased glycolysis in PS2-deficient cells (PS2KO). This phenotype was restored by the stable re-expression of human PS2. Still, all these results were obtained in immortalized cells, and one bottom-line question is to know whether these observations hold true in central nervous system (CNS) cells. To that end, we carried out primary cultures of PS1 knockdown (KD), PS2KO, and PS1KD/PS2KO (PSdKO) neurons and astrocytes. They were obtained from the same litter by crossing PS2 heterozygous; PS1 floxed (PS2+/-; PS1flox/flox) animals. Genetic downregulation of PS1 was achieved by lentiviral expression of the Cre recombinase in primary cultures. Strikingly, we did not observe any mitochondrial phenotype in PS1KD, PS2KO, or PSdKO primary cultures in basal conditions. Mitochondrial respiration and membrane potential were similar in all models, as were the glycolytic flux and NAD+/NADH ratio. Likewise, mitochondrial morphology and content was unaltered by PS expression. We further investigated the differences between results we obtained here in primary nerve cells and those previously reported in MEF cell lines by analyzing PS2KO primary fibroblasts. We found no mitochondrial dysfunction in this model, in line with observations in PS2KO primary neurons and astrocytes. Together, our results indicate that the mitochondrial phenotype observed in immortalized PS2-deficient cell lines cannot be extrapolated to primary neurons, astrocytes, and even to primary fibroblasts. The PS-dependent mitochondrial phenotype reported so far might therefore be the consequence of a cell immortalization process and should be critically reconsidered regarding its relevance to AD.

15.
Life Sci Alliance ; 2(2)2019 04.
Article in English | MEDLINE | ID: mdl-30894406

ABSTRACT

Mechanisms driving cognitive improvements following nuclear receptor activation are poorly understood. The peroxisome proliferator-activated nuclear receptor alpha (PPARα) forms heterodimers with the nuclear retinoid X receptor (RXR). We report that PPARα mediates the improvement of hippocampal synaptic plasticity upon RXR activation in a transgenic mouse model with cognitive deficits. This improvement results from an increase in GluA1 subunit expression of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor, eliciting an AMPA response at the excitatory synapses. Associated with a two times higher PPARα expression in males than in females, we show that male, but not female, PPARα null mutants display impaired hippocampal long-term potentiation. Moreover, PPARα knockdown in the hippocampus of cognition-impaired mice compromises the beneficial effects of RXR activation on synaptic plasticity only in males. Furthermore, selective PPARα activation with pemafibrate improves synaptic plasticity in male cognition-impaired mice, but not in females. We conclude that striking sex differences in hippocampal synaptic plasticity are observed in mice, related to differences in PPARα expression levels.


Subject(s)
Gene Dosage/genetics , Long-Term Potentiation/genetics , Neuronal Plasticity/genetics , PPAR alpha/genetics , PPAR alpha/metabolism , Animals , Benzoxazoles/pharmacology , Butyrates/pharmacology , Cells, Cultured , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Female , Gene Knockdown Techniques , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Male , Mice , Mice, Transgenic , PPAR alpha/agonists , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Retinoid X Receptors/metabolism , Sex Factors , Signal Transduction/drug effects
16.
J Biol Chem ; 294(15): 5854-5866, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30755484

ABSTRACT

Extracellular deposition of ß-amyloid (Aß) peptides in the brain is a hallmark of Alzheimer's disease (AD). Upon ß-secretase-mediated cleavage of the ß C-terminal fragment (ß-CTF) from the Aß precursor protein, the γ-secretase complex produces the Aß peptides associated with AD. The familial T43I mutation within the transmembrane domain of the ß-CTF (also referred to as C99) increases the ratio between the Aß42 and Aß40 peptides largely due to a decrease in Aß40 formation. Aß42 is the principal component of amyloid deposits within the brain parenchyma, and an increase in the Aß42/Aß40 ratio is correlated with early-onset AD. Using NMR and FTIR spectroscopy, here we addressed how the T43I substitution influences the structure of C55, the minimal sequence containing the entire extracellular and transmembrane (TM) domains of C99 needed for γ-secretase processing. 13C NMR chemical shifts indicated that the T43I substitution increases helical structure within the TM domain of C55. These structural changes were associated with a shift of the C55 dimer to the monomer and an increase in the tilt of the TM helix relative to the membrane normal in the T43I mutant compared with that of WT C55. The A21G (Flemish) mutation was previously found to increase secreted Aß40 levels; here, we combined this mutation in the extracellular domain of C99 with T43I and observed that the T43I/A21G double mutant decreases Aß40 formation. We discuss how the observed structural changes in the T43I mutant may decrease Aß40 formation and increase the Aß42/Aß40 ratio.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/chemistry , Mutation, Missense , Peptide Fragments/chemistry , Peptides/chemistry , Amino Acid Substitution , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptides/genetics , Peptides/metabolism , Protein Domains
17.
Front Cell Neurosci ; 12: 435, 2018.
Article in English | MEDLINE | ID: mdl-30524243

ABSTRACT

Aß peptides, the major components of Alzheimer's disease (AD) amyloid deposits, are released following sequential cleavages by secretases of its precursor named the amyloid precursor protein (APP). In addition to secretases, degradation pathways, in particular the endosomal/lysosomal and proteasomal systems have been reported to contribute to APP processing. However, the respective role of each of these pathways toward APP metabolism remains to be established. To address this, we used HEK 293 cells and primary neurons expressing full-length wild type APP or the ß-secretase-derived C99 fragment (ß-CTF) in which degradation pathways were selectively blocked using pharmacological drugs. APP metabolites, including carboxy-terminal fragments (CTFs), soluble APP (sAPP) and Aß peptides were studied. In this report, we show that APP-CTFs produced from endogenous or overexpressed full-length APP are mainly processed by γ-secretase and the endosomal/lysosomal pathway, while in sharp contrast, overexpressed C99 is mainly degraded by the proteasome and to a lesser extent by γ-secretase.

18.
Front Neurol ; 9: 384, 2018.
Article in English | MEDLINE | ID: mdl-29899726

ABSTRACT

The current inability of clinical criteria to accurately identify the "at-risk group" for Amyotrophic Lateral Sclerosis (ALS) development as well as its unknown etiology are fueling the interest in biomarkers aimed at completing clinical approaches for the diagnosis. The Glial cell line-derived neurotrophic factor (GDNF) is a diffusible peptide critically involved in neuronal differentiation and survival. GDNF is largely studied in various neurological and neuromuscular diseases, with a great interest in the peripheral nervous system (PNS). The recent discovery of Amyloid Precursor Protein (APP)-dependent GDNF regulation driving neuro-muscular junctions' formation in APP null transgenic mice, prompts to study whether neurodegeneration relies on loss or gain of APP function and suggests that it could affect peripheral processes. Here, we explored a brand-new aspect of the loss of trophic support in ALS by measuring GDNF, APP, soluble APP fragments and Aß peptides levels in SOD1WT or SOD1G93A transgenic mouse models of ALS and in human biological fluids [i.e. serum and cerebrospinal fluid (CSF)] from ALS patients and control subjects. Our results show that both GDNF and soluble APP fragments levels are altered at the onset of motor deficits in mice and that their levels are also modified in patient samples. This study indicates that both GDNF and soluble APPα represent possible biomarkers for ALS.

19.
J Cell Mol Med ; 22(2): 823-833, 2018 02.
Article in English | MEDLINE | ID: mdl-28994238

ABSTRACT

The two presenilin-1 (PS1) and presenilin-2 (PS2) homologs are the catalytic core of the γ-secretase complex, which has a major role in cell fate decision and Alzheimer's disease (AD) progression. Understanding the precise contribution of PS1- and PS2-dependent γ-secretases to the production of ß-amyloid peptide (Aß) from amyloid precursor protein (APP) remains an important challenge to design molecules efficiently modulating Aß release without affecting the processing of other γ-secretase substrates. To that end, we studied PS1- and PS2-dependent substrate processing in murine cells lacking presenilins (PSs) (PS1KO, PS2KO or PS1-PS2 double-KO noted PSdKO) or stably re-expressing human PS1 or PS2 in an endogenous PS-null (PSdKO) background. We characterized the processing of APP and Notch on both endogenous and exogenous substrates, and we investigated the effect of pharmacological inhibitors targeting the PSs activity (DAPT and L-685,458). We found that murine PS1 γ-secretase plays a predominant role in APP and Notch processing when compared to murine PS2 γ-secretase. The inhibitors blocked more efficiently murine PS2- than murine PS1-dependent processing. Human PSs, especially human PS1, expression in a PS-null background efficiently restored APP and Notch processing. Strikingly, and contrary to the results obtained on murine PSs, pharmacological inhibitors appear to preferentially target human PS1- than human PS2-dependent γ-secretase activity.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Presenilin-1/metabolism , Presenilin-2/metabolism , Amyloid beta-Peptides/metabolism , Animals , Biocatalysis , Fibroblasts/metabolism , Humans , Mice, Knockout , Receptors, Notch/metabolism , Substrate Specificity
20.
Sci Rep ; 7(1): 17159, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215043

ABSTRACT

Familial mutations in C99 can increase the total level of the soluble Aß peptides produced by proteolysis, as well as the Aß42/Aß40 ratio, both of which are linked to the progression of Alzheimer's disease. We show that the extracellular sequence of C99 forms ß-sheet structure upon interaction with membrane bilayers. Mutations that disrupt this structure result in a significant increase in Aß production and, in specific cases, result in an increase in the amount of Aß42 relative to Aß40. Fourier transform infrared and solid-state NMR spectroscopic studies reveal a central ß-hairpin within the extracellular sequence comprising Y10-E11-V12 and L17-V18-F19 connected by a loop involving H13-H14-Q15. These results suggest how familial mutations in the extracellular sequence influence C99 processing and provide a structural basis for the development of small molecule modulators that would reduce Aß production.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Amyloid/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation, beta-Strand , Amyloid/chemistry , Humans , Models, Molecular , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...