15 research outputs found

    Investigating the role of presenilins in mitochondrial activity and metabolism

    No full text
    Presenilines (PS1 & PS2) are known for their involvement in the production of the amyloid β peptide (Aβ) which accumulates and forms the characteristic senile plaques of Alzheimer's disease (AD). In addition to this role, the PSs have been assigned numerous physiological functions. They are enriched in the lipid rafts present at the plasma membrane but also in the sub-compartments called mitochondrial-associated membrane (MAM) where the reticulum and the mitochondria interact to regulate various processes. In this work, we have studied the role of PSs in cell metabolism with a particular interest in mitochondria and lipid metabolism. Preliminary data obtained from fibroblastic lines deficient in PSs show that PSs could be involved in endocytosis and extracellular vesicles formation. These processes are currently widely studied for their potential role in the spread of Aβ. We also observed a PS2-dependent phenotype only in these cell lines. Indeed, the activity and expression of the electron transport chain was deficient, as was the NAD+/NADH ratio. Anaerobic glycolysis was increased in order to compensate energy needs of the cell. However, no mitochondrial deficit was observed in primary neuronal, astrocytic and fibroblast cultures. Immortalization of the cell lines mimics the uncontrolled proliferation of cancer cells, which may indicate a contribution of PSs to cancer-related cellular processes. Nevertheless, these results question the contribution of PSs in the bioenergetic aspect of AD and raise questions about these widely used lines.(BIFA - Sciences biomédicales et pharmaceutiques) -- UCL, 202

    Presenilin-Deficient Neurons and Astrocytes Display Normal Mitochondrial Phenotypes

    No full text
    Presenilin 1 (PS1) and Presenilin 2 (PS2) are predominantly known as the catalytic subunits of the γ-secretase complex that generates the amyloid-β (Aβ) 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

    APP-dependent glial cell line-derived neurotrophic factor gene expression drives neuromuscular junction formation.

    No full text
    Besides its crucial role in the pathogenesis of Alzheimer's disease, the knowledge of amyloid precursor protein (APP) physiologic functions remains surprisingly scarce. Here, we show that APP regulates the transcription of the glial cell line-derived neurotrophic factor (GDNF). APP-dependent regulation of GDNF expression affects muscle strength, muscular trophy, and both neuronal and muscular differentiation fundamental for neuromuscular junction (NMJ) maturation in vivo. In a nerve-muscle coculture model set up to modelize NMJ formation in vitro, silencing of muscular APP induces a 30% decrease in secreted GDNF levels and a 40% decrease in the total number of NMJs together with a significant reduction in the density of acetylcholine vesicles at the presynaptic site and in neuronal maturation. These defects are rescued by GDNF expression in muscle cells in the conditions where muscular APP has been previously silenced. Expression of GDNF in muscles of amyloid precursor protein null mice corrected the aberrant synaptic morphology of NMJs. Our findings highlight for the first time that APP-dependent GDNF expression drives the process of NMJ formation, providing new insights into the link between APP gene regulatory network and physiologic functions

    Mechanism of Cellular Formation and In Vivo Seeding Effects of Hexameric β-Amyloid Assemblies.

    No full text
    The β-amyloid peptide (Aβ) is found as amyloid fibrils in senile plaques, a typical hallmark of Alzheimer's disease (AD). However, intermediate soluble oligomers of Aβ are now recognized as initiators of the pathogenic cascade leading to AD. Studies using recombinant Aβ have shown that hexameric Aβ in particular acts as a critical nucleus for Aβ self-assembly. We recently isolated hexameric Aβ assemblies from a cellular model, and demonstrated their ability to enhance Aβ aggregation in vitro. Here, we report the presence of similar hexameric-like Aβ 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 Aβ. 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 Aβ assemblies in extracellular vesicles. Further, we assessed the contribution of hexameric Aβ to the development of amyloid pathology. We report the early presence of hexameric-like Aβ assemblies in both transgenic mice brains exhibiting human Aβ pathology and in the cerebrospinal fluid of AD patients, suggesting hexameric Aβ as a potential early AD biomarker. Finally, cell-derived hexameric Aβ was found to seed other human Aβ forms, resulting in the aggravation of amyloid deposition in vivo and neuronal toxicity in vitro

    APP-dependent regulation of GDNF expression and its involvement in neuromuscular junction

    No full text
    OBJECTIVES: Amyloid precursor protein (APP) plays a crucial role in Alzheimer's disease (AD) pathogenesis since its proteolytical cleavage generates β-amyloid peptide, the major constituent of senile plaques. However, APP precise physiological role has not been clarified yet. Interestingly, we observed that APP regulates the transcription of GDNF (Glial cell-line Derived Neutrophic Factor) in cell lines and in transgenic mice. The aim of our work is to investigate this novel APP function. METHODS: Following microarray analysis, quantitative RT-PCR, ELISA and reporter gene assays were performed on mouse embryonic fibroblast cell line (MEF) knock-out for APP. APP expression was monitored during myotube differentiation in C2C12 mouse myoblast has packed or not with a GDNF expression plasmid. Studies on APP knock-out (APP KO) mice included grip strength tests, mechanic measurements on isolated extensor digitorum longus (EDL) and immunohistochemistry (IHC) to investigate muscular atrophy and neuromuscular junctions (NMJs) formation. RESULTS: We found that GDNF mRNA and protein levels together with GDNF promoter transcriptional activity are downregulated in APP KO MEFs compared to control cells. In C2C12, we observed that consequent to myotube differentiation GDNF and APP protein levels increase. Moreover transfection of the GDNF-encoding plasmid favors myotube formation and correlates with APP increase. We showed that APP KO mice muscular phenotype and defective neuromuscular synapses are associated with GDNF downregulation. CONCLUSIONS: Our findings highlight for the first time a role for APP in the regulation of GDNF expression with implications in muscular differentiation and NMJs formation and function

    Presenilin 2-Dependent Maintenance of Mitochondrial Oxidative Capacity and Morphology.

    Get PDF
    Mitochondrial dysfunction plays a pivotal role in the progression of Alzheimer's disease (AD), and yet the mechanisms underlying the impairment of mitochondrial function in AD remain elusive. Recent evidence suggested a role for Presenilins (PS1 or PS2) in mitochondrial function. Mutations of PSs, the catalytic subunits of the γ-secretase complex, are responsible for the majority of inherited AD cases (FAD). PSs were shown to be present in mitochondria and particularly enriched in mitochondria-associated membranes (MAM), where PS2 is involved in the calcium shuttling between mitochondria and the endoplasmic reticulum (ER). We investigated the precise contribution of PS1 and PS2 to the bioenergetics of the cell and to mitochondrial morphology in cell lines derived from wild type (PS+/+), PS1/2 double knock-out (PSdKO), PS2KO and PS1KO embryos. Our results showed a significant impairment in the respiratory capacity of PSdKO and PS2KO cells with reduction of basal oxygen consumption, oxygen utilization dedicated to ATP production and spare respiratory capacity. In line with these functional defects, we found a decrease in the expression of subunits responsible for mitochondrial oxidative phosphorylation (OXPHOS) associated with an altered morphology of the mitochondrial cristae. This OXPHOS disruption was accompanied by a reduction of the NAD(+)/NADH ratio. Still, neither ADP/ATP ratio nor mitochondrial membrane potential (ΔΨ) were affected, suggesting the existence of a compensatory mechanism for energetic balance. We observed indeed an increase in glycolytic flux in PSdKO and PS2KO cells. All these effects were truly dependent on PS2 since its stable re-expression in a PS2KO background led to a complete restoration of the parameters impaired in the absence of PS2. Our data clearly demonstrate here the crucial role of PS2 in mitochondrial function and cellular bioenergetics, pointing toward its peculiar role in the formation and integrity of the electron transport chain

    APP-dependent glial cell line-derived neurotrophic factor gene expression drives neuromuscular junction formation.

    Get PDF
    Besides its crucial role in the pathogenesis of Alzheimer's disease, the knowledge of amyloid precursor protein (APP) physiologic functions remains surprisingly scarce. Here, we show that APP regulates the transcription of the glial cell line-derived neurotrophic factor (GDNF). APP-dependent regulation of GDNF expression affects muscle strength, muscular trophy, and both neuronal and muscular differentiation fundamental for neuromuscular junction (NMJ) maturation in vivo In a nerve-muscle coculture model set up to modelize NMJ formation in vitro, silencing of muscular APP induces a 30% decrease in secreted GDNF levels and a 40% decrease in the total number of NMJs together with a significant reduction in the density of acetylcholine vesicles at the presynaptic site and in neuronal maturation. These defects are rescued by GDNF expression in muscle cells in the conditions where muscular APP has been previously silenced. Expression of GDNF in muscles of amyloid precursor protein null mice corrected the aberrant synaptic morphology of NMJs. Our findings highlight for the first time that APP-dependent GDNF expression drives the process of NMJ formation, providing new insights into the link between APP gene regulatory network and physiologic functions.-Stanga, S., Zanou, N., Audouard, E., Tasiaux, B., Contino, S., Vandermeulen, G., René, F., Loeffler, J.-P., Clotman, F., Gailly, P., Dewachter, I., Octave, J.-N., Kienlen-Campard, P. APP-dependent glial cell line-derived neurotrophic factor gene expression drives neuromuscular junction formation
    corecore