Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
NPJ Parkinsons Dis ; 8(1): 52, 2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35468903

ABSTRACT

Lipid profiles in biological fluids from patients with Parkinson's disease (PD) are increasingly investigated in search of biomarkers. However, the lipid profiles in genetic PD remain to be determined, a gap of knowledge of particular interest in PD associated with mutant α-synuclein (SNCA), given the known relationship between this protein and lipids. The objective of this research is to identify serum lipid composition from SNCA A53T mutation carriers and to compare these alterations to those found in cells and transgenic mice carrying the same genetic mutation. We conducted an unbiased lipidomic analysis of 530 lipid species from 34 lipid classes in serum of 30 participants with SNCA mutation with and without PD and 30 healthy controls. The primary analysis was done between 22 PD patients with SNCA+ (SNCA+/PD+) and 30 controls using machine-learning algorithms and traditional statistics. We also analyzed the lipid composition of human clonal-cell lines and tissue from transgenic mice overexpressing the same SNCA mutation. We identified specific lipid classes that best discriminate between SNCA+/PD+ patients and healthy controls and found certain lipid species, mainly from the glycerophosphatidylcholine and triradylglycerol classes, that are most contributory to this discrimination. Most of these alterations were also present in human derived cells and transgenic mice carrying the same mutation. Our combination of lipidomic and machine learning analyses revealed alterations in glycerophosphatidylcholine and triradylglycerol in sera from PD patients as well as cells and tissues expressing mutant α-Syn. Further investigations are needed to establish the pathogenic significance of these α-Syn-associated lipid changes.

2.
J Parkinsons Dis ; 11(3): 1141-1155, 2021.
Article in English | MEDLINE | ID: mdl-33814463

ABSTRACT

BACKGROUND: The role of the lipidome as a biomarker for Parkinson's disease (PD) is a relatively new field that currently only focuses on PD diagnosis. OBJECTIVE: To identify a relevant lipidome signature for PD severity markers. METHODS: Disease severity of 149 PD patients was assessed by the Unified Parkinson's Disease Rating Scale (UPDRS) and the Montreal Cognitive Assessment (MoCA). The lipid composition of whole blood samples was analyzed, consisting of 517 lipid species from 37 classes; these included all major classes of glycerophospholipids, sphingolipids, glycerolipids, and sterols. To handle the high number of lipids, the selection of lipid species and classes was consolidated via analysis of interrelations between lipidomics and disease severity prediction using the random forest machine-learning algorithm aided by conventional statistical methods. RESULTS: Specific lipid classes dihydrosphingomyelin (dhSM), plasmalogen phosphatidylethanolamine (PEp), glucosylceramide (GlcCer), dihydro globotriaosylceramide (dhGB3), and to a lesser degree dihydro GM3 ganglioside (dhGM3), as well as species dhSM(20:0), PEp(38:6), PEp(42:7), GlcCer(16:0), GlcCer(24:1), dhGM3(22:0), dhGM3(16:0), and dhGB3(16:0) contribute to PD severity prediction of UPDRS III score. These, together with age, age at onset, and disease duration, also contribute to prediction of UPDRS total score. We demonstrate that certain lipid classes and species interrelate differently with the degree of severity of motor symptoms between men and women, and that predicting intermediate disease stages is more accurate than predicting less or more severe stages. CONCLUSION: Using machine-learning algorithms and methodologies, we identified lipid signatures that enable prediction of motor severity in PD. Future studies should focus on identifying the biological mechanisms linking GlcCer, dhGB3, dhSM, and PEp with PD severity.


Subject(s)
Lipidomics , Parkinson Disease , Biomarkers , Female , Humans , Lipids , Machine Learning , Male , Parkinson Disease/diagnosis , Severity of Illness Index
3.
EMBO J ; 39(20): e103791, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32865299

ABSTRACT

The link between cholesterol homeostasis and cleavage of the amyloid precursor protein (APP), and how this relationship relates to Alzheimer's disease (AD) pathogenesis, is still unknown. Cellular cholesterol levels are regulated through crosstalk between the plasma membrane (PM), where most cellular cholesterol resides, and the endoplasmic reticulum (ER), where the protein machinery that regulates cholesterol levels resides. The intracellular transport of cholesterol from the PM to the ER is believed to be activated by a lipid-sensing peptide(s) in the ER that can cluster PM-derived cholesterol into transient detergent-resistant membrane domains (DRMs) within the ER, also called the ER regulatory pool of cholesterol. When formed, these cholesterol-rich domains in the ER maintain cellular homeostasis by inducing cholesterol esterification as a mechanism of detoxification while attenuating its de novo synthesis. In this manuscript, we propose that the 99-aa C-terminal fragment of APP (C99), when delivered to the ER for cleavage by γ-secretase, acts as a lipid-sensing peptide that forms regulatory DRMs in the ER, called mitochondria-associated ER membranes (MAM). Our data in cellular AD models indicates that increased levels of uncleaved C99 in the ER, an early phenotype of the disease, upregulates the formation of these transient DRMs by inducing the internalization of extracellular cholesterol and its trafficking from the PM to the ER. These results suggest a novel role for C99 as a mediator of cholesterol disturbances in AD, potentially explaining early hallmarks of the disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Animals , Cell Line , Cholesterol/biosynthesis , Endoplasmic Reticulum/genetics , Fibroblasts/metabolism , Gene Knockdown Techniques , Gene Silencing , Humans , Induced Pluripotent Stem Cells , Lipid Metabolism , Lipidomics , Mice , Mitochondria/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/genetics , Presenilin-2/metabolism , Protein Domains , RNA, Small Interfering , Sphingomyelin Phosphodiesterase/metabolism
4.
Curr Opin Clin Nutr Metab Care ; 23(2): 68-75, 2020 03.
Article in English | MEDLINE | ID: mdl-32004239

ABSTRACT

PURPOSE OF REVIEW: The purpose of this brief review is to gain an understanding on the multiple roles that lipids exert on the brain, and to highlight new ideas in the impact of lipid homeostasis in the regulation of synaptic transmission. RECENT FINDINGS: Recent data underline the crucial function of lipid homeostasis in maintaining neuronal function and synaptic plasticity. Moreover, new advances in analytical approaches to study lipid classes and species is opening a new door to understand and monitor how alterations in lipid pathways could shed new light into the pathogenesis of neurodegeneration. SUMMARY: Lipids are one of the most essential elements of the brain. However, our understanding of the role of lipids within the central nervous system is still largely unknown. Identifying the molecular mechanism (s) by which lipids can regulate neuronal transmission represents the next frontier in neuroscience, and a new challenge in our understanding of the brain and the mechanism(s) behind neurological disorders.


Subject(s)
Brain/metabolism , Lipid Metabolism/physiology , Lipids/physiology , Nervous System Diseases/metabolism , Neurons/metabolism , Animals , Homeostasis , Humans , Neuronal Plasticity
5.
J Neurosci ; 39(36): 7074-7085, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31300519

ABSTRACT

Maintaining a pool of functional mitochondria requires degradation of damaged ones within the cell. PINK1 is critical in this quality-control process: loss of mitochondrial membrane potential causes PINK1 to accumulate on the mitochondrial surface, triggering mitophagy. However, little is known about how PINK1 is regulated. Recently, we showed that PINK1 content is kept low in healthy mitochondria by continuous ubiquitination and proteasomal degradation of its mature form via a mechanism inconsistent with the proposed N-end rule process. Using both human female and monkey cell lines, we now demonstrate that once generated within the mitochondria, 52 kDa PINK1 adopts a mitochondrial topology most consistent with it being at the mitochondrial-endoplasmic reticulum (ER) interface. From this particular submitochondrial location, PINK1 interacts with components of the ER-associated degradation pathway, such as the E3 ligases gp78 and HRD1, which cooperate to catalyze PINK1 ubiquitination. The valosin-containing protein and its cofactor, UFD1, then target ubiquitinated PINK1 for proteasomal degradation. Our data show that PINK1 in healthy mitochondria is negatively regulated via an interplay between mitochondria and ER, and shed light on how this mitochondrial protein gains access to the proteasome.SIGNIFICANCE STATEMENT Regulation of mitochondrial content of PINK1, a contributor to mitophagy, is an important area of research. Recently, we found that PINK1 content is kept low in healthy mitochondria by continuous ubiquitination and proteasomal degradation. We now extend and refine this novel finding by showing that PINK1 localizes at the mitochondrial-endoplasmic reticulum (ER) interface, from where it interacts with the ER-associated degradation machinery, which catalyzes its ubiquitination and transfer to the proteasome. Thus, these data show that PINK1 in healthy mitochondria is negatively regulated via a mitochondria and ER interplay, and how this mitochondrial protein gains access to the proteasome.


Subject(s)
Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Protein Kinases/metabolism , Proteolysis , Ubiquitination , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Receptors, Autocrine Motility Factor/metabolism , Ubiquitin-Protein Ligases/metabolism , Valosin Containing Protein/metabolism
6.
J Clin Invest ; 129(1): 34-45, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30601141

ABSTRACT

Mitochondrial respiratory deficiencies have been observed in numerous neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. For decades, these reductions in oxidative phosphorylation (OxPhos) have been presumed to trigger an overall bioenergetic crisis in the neuron, resulting in cell death. While the connection between respiratory defects and neuronal death has never been proven, this hypothesis has been supported by the detection of nonspecific mitochondrial DNA mutations in these disorders. These findings led to the notion that mitochondrial respiratory defects could be initiators of these common neurodegenerative disorders, instead of being consequences of a prior insult, a theory we believe to be misconstrued. Herein, we review the roots of this mitochondrial hypothesis and offer a new perspective wherein mitochondria are analyzed not only from the OxPhos point of view, but also as a complex organelle residing at the epicenter of many metabolic pathways.


Subject(s)
Alzheimer Disease/metabolism , Mitochondria/metabolism , Models, Neurological , Neurons/metabolism , Oxidative Phosphorylation , Parkinson Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cell Death , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Humans , Mitochondria/genetics , Mitochondria/pathology , Mutation , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology
7.
Hum Mol Genet ; 28(11): 1782-1800, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30649465

ABSTRACT

Charcot-Marie-Tooth disease (CMT) type 2A is a form of peripheral neuropathy, due almost exclusively to dominant mutations in the nuclear gene encoding the mitochondrial protein mitofusin-2 (MFN2). However, there is no understanding of the relationship of clinical phenotype to genotype. MFN2 has two functions: it promotes inter-mitochondrial fusion and mediates endoplasmic reticulum (ER)-mitochondrial tethering at mitochondria-associated ER membranes (MAM). MAM regulates a number of key cellular functions, including lipid and calcium homeostasis, and mitochondrial behavior. To date, no studies have been performed to address whether mutations in MFN2 in CMT2A patient cells affect MAM function, which might provide insight into pathogenesis. Using fibroblasts from three CMT2AMFN2 patients with different mutations in MFN2, we found that some, but not all, examined aspects of ER-mitochondrial connectivity and of MAM function were indeed altered, and correlated with disease severity. Notably, however, respiratory chain function in those cells was unimpaired. Our results suggest that CMT2AMFN2 is a MAM-related disorder but is not a respiratory chain-deficiency disease. The alterations in MAM function described here could also provide insight into the pathogenesis of other forms of CMT.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Endoplasmic Reticulum/genetics , GTP Phosphohydrolases/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Adult , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Endoplasmic Reticulum/metabolism , Energy Metabolism/genetics , Female , Fibroblasts/metabolism , Genotype , Humans , Male , Middle Aged , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Membranes/metabolism , Mutation , Oxidative Phosphorylation , Severity of Illness Index
8.
EMBO J ; 36(22): 3356-3371, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29018038

ABSTRACT

In the amyloidogenic pathway associated with Alzheimer disease (AD), the amyloid precursor protein (APP) is cleaved by ß-secretase to generate a 99-aa C-terminal fragment (C99) that is then cleaved by γ-secretase to generate the ß-amyloid (Aß) found in senile plaques. In previous reports, we and others have shown that γ-secretase activity is enriched in mitochondria-associated endoplasmic reticulum (ER) membranes (MAM) and that ER-mitochondrial connectivity and MAM function are upregulated in AD We now show that C99, in addition to its localization in endosomes, can also be found in MAM, where it is normally processed rapidly by γ-secretase. In cell models of AD, however, the concentration of unprocessed C99 increases in MAM regions, resulting in elevated sphingolipid turnover and an altered lipid composition of both MAM and mitochondrial membranes. In turn, this change in mitochondrial membrane composition interferes with the proper assembly and activity of mitochondrial respiratory supercomplexes, thereby likely contributing to the bioenergetic defects characteristic of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Mitochondria/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Cell Respiration , Endoplasmic Reticulum/ultrastructure , Humans , Intracellular Membranes/ultrastructure , Mice , Mitochondria/ultrastructure , Mutation/genetics , Oxygen Consumption , Presenilins/genetics , Protein Transport , Sphingolipids/metabolism , Up-Regulation
9.
Cell Rep ; 20(1): 30-39, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28683321

ABSTRACT

Along with Parkin, PINK1 plays a critical role in maintaining mitochondrial quality control. Although PINK1 is expressed constitutively, its level is kept low in healthy mitochondria by polyubiquitination and ensuing proteasomal degradation of its mature, 52 kDa, form. We show here that the target of PINK1 polyubiquitination is the mature form and is mediated by ubiquitination of a conserved lysine at position 137. Notably, the full-length protein also contains Lys-137 but is not ubiquitinated. On the basis of our data, we propose that cleavage of full-length PINK1 at Phe-104 disrupts the major hydrophobic membrane-spanning domain in the protein, inducing a conformation change in the resultant mature form that exposes Lys-137 to the cytosol for subsequent modification by the ubiquitination machinery. Thus, the balance between the full-length and mature PINK1 allows its levels to be regulated via ubiquitination of the mature form and ensures that PINK1 functions as a mitochondrial quality control factor.


Subject(s)
Protein Kinases/metabolism , Ubiquitination , Amino Acid Motifs , Animals , Cell Line , Conserved Sequence , Mice , Protein Domains , Protein Kinases/chemistry , Protein Kinases/genetics
10.
EMBO Rep ; 17(1): 27-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26564908

ABSTRACT

In addition to the appearance of senile plaques and neurofibrillary tangles, Alzheimer's disease (AD) is characterized by aberrant lipid metabolism and early mitochondrial dysfunction. We recently showed that there was increased functionality of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a subdomain of the ER involved in lipid and cholesterol homeostasis, in presenilin-deficient cells and in fibroblasts from familial and sporadic AD patients. Individuals carrying the ε4 allele of apolipoprotein E (ApoE4) are at increased risk for developing AD compared to those carrying ApoE3. While the reason for this increased risk is unknown, we hypothesized that it might be associated with elevated MAM function. Using an astrocyte-conditioned media (ACM) model, we now show that ER-mitochondrial communication and MAM function-as measured by the synthesis of phospholipids and of cholesteryl esters, respectively-are increased significantly in cells treated with ApoE4-containing ACM as compared to those treated with ApoE3-containing ACM. Notably, this effect was seen with lipoprotein-enriched preparations, but not with lipid-free ApoE protein. These data are consistent with a role of upregulated MAM function in the pathogenesis of AD and may help explain, in part, the contribution of ApoE4 as a risk factor in the disease.


Subject(s)
Apolipoprotein E4/metabolism , Astrocytes/physiology , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Mitochondria/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Animals , Apolipoprotein E4/chemistry , Apolipoprotein E4/genetics , Cholesterol/metabolism , Cholesterol Esters/biosynthesis , Culture Media, Conditioned/chemistry , Endoplasmic Reticulum/genetics , Humans , Lipid Metabolism , Lipoproteins/metabolism , Mice , Phospholipids/biosynthesis , Transcriptional Activation , Up-Regulation
11.
Mov Disord ; 30(8): 1026-33, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25952565

ABSTRACT

Familial cases of Parkinson's disease (PD) can be associated with overexpression or mutation of α-synuclein, a synaptic protein reported to be localized mainly in the cytosol and mitochondria. We recently showed that wild-type α-synuclein is not present in mitochondria, as previously thought, but rather is located in mitochondrial-associated endoplasmic reticulum membranes. Remarkably, we also found that PD-related mutated α-synuclein results in its reduced association with mitochondria-associated membranes, coincident with a lower degree of apposition of endoplasmic reticulum with mitochondria and an increase in mitochondrial fragmentation, as compared with wild-type. This new subcellular localization of α-synuclein raises fundamental questions regarding the relationship of α-synuclein to mitochondria-associated membranes function, in both normal and pathological states. In this article, we attempt to relate aspects of PD pathogenesis to what is known about mitochondria-associated membranes' behavior and function. We hypothesize that early events occurring in dopaminergic neurons at the level of the mitochondria-associated membranes could cause long-term disturbances that lead to PD.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Mitochondria/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Humans
12.
Front Neuroanat ; 9: 17, 2015.
Article in English | MEDLINE | ID: mdl-25755636

ABSTRACT

α-synuclein (α-syn) is one of the genes that when mutated or overexpressed causes Parkinson's Disease (PD). Initially, it was described as a synaptic terminal protein and later was found to be localized at mitochondria. Mitochondria-associated membranes (MAM) have emerged as a central endoplasmic reticulum (ER) subcellular compartments where key functions of the cell occur. These domains, enriched in cholesterol and anionic phospholipids, are where calcium homeostasis, lipid transfer, and cholesterol metabolism are regulated. Some proteins, related to mitochondrial dynamics and function, are also localized to this area. Several neurodegenerative diseases have shown alterations in MAM functions and resident proteins, including Charcot Marie-Tooth and Alzheimer's disease (AD). We have recently reported that MAM function is downregulated in cell and mouse models of PD expressing pathogenic mutations of α-syn. This review focuses on the possible role of α-syn in these cellular domains and the early pathogenic features of PD that could be explained by α-syn-MAM disturbances.

13.
Stroke ; 45(2): 413-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24385273

ABSTRACT

BACKGROUND AND PURPOSE: It has been proposed that the deposition of the ß-amyloid peptide (Aß) in the brain parenchyma and brain blood vessels has deleterious effects. We tested the hypothesis that the levels of plasma Aß are related to the outcome in patients with intracerebral hemorrhage. METHODS: In a multicenter study, we prospectively included patients with spontaneous intracerebral hemorrhage within the first 24 hours after onset. At admission, we measured plasma Aß40 and Aß42 levels using ELISA techniques. Also, we recorded age, sex, vascular risk factors, National Institutes of Health Stroke Scale score, presence of intraventricular hemorrhage, localization, cause, and volume of the hematoma. We obtained the modified Rankin scale and defined a unfavorable outcome as modified Rankin scale >2 at 3 months. Bivariate and multivariate regression analyses were performed. RESULTS: We studied 160 patients (mean age, 73.8±11.3 years; 59.4% of them were men). A favorable outcome was observed in 64 (40%) of the patients. In the bivariate analyses, unfavorable outcome was associated with high age, female sex, diabetes mellitus, presence of intraventricular hemorrhage, high blood glucose, high National Institutes of Health Stroke Scale score, high volume, and high plasma levels of Aß42 and Aß40. The multivariate analysis showed that increased age (odds ratio, 1.07; 95% confidence interval, 1.035-1.21; P<0.0001), high admission National Institutes of Health Stroke Scale score (odds ratio, 1.29, 95% confidence interval, 1.17-1.42; P<0.0001), presence of diabetes mellitus (odds ratio, 4.15; 95% confidence interval, 1.21-14.1; P=0.02), and Aß42 levels >9.7 pg/mL (odds ratio, 4.11; 95% confidence interval, 1.65-10.1; P=0.02) were independently associated with an increased likelihood of an unfavorable outcome. CONCLUSIONS: High levels of plasma Aß42 in patients with acute intracerebral hemorrhage are associated with a poor functional prognosis.


Subject(s)
Amyloid beta-Peptides/blood , Cerebral Hemorrhage/blood , Age Factors , Aged , Aged, 80 and over , Confidence Intervals , Data Interpretation, Statistical , Diabetes Complications/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Peptide Fragments/blood , Prognosis , Prospective Studies , Regression Analysis , Sex Factors , Treatment Outcome
14.
J Neurosci ; 34(1): 249-59, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24381286

ABSTRACT

Familial Parkinson disease is associated with mutations in α-synuclein (α-syn), a presynaptic protein that has been localized not only to the cytosol, but also to mitochondria. We report here that wild-type α-syn from cell lines, and brain tissue from humans and mice, is present not in mitochondria but rather in mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a structurally and functionally distinct subdomain of the ER. Remarkably, we found that pathogenic point mutations in human α-syn result in its reduced association with MAM, coincident with a lower degree of apposition of ER with mitochondria, a decrease in MAM function, and an increase in mitochondrial fragmentation compared with wild-type. Although overexpression of wild-type α-syn in mutant α-syn-expressing cells reverted the fragmentation phenotype, neither overexpression of the mitochondrial fusion/MAM-tethering protein MFN2 nor inhibition/ablation of the mitochondrial fission protein DRP1 was able to do so, implying that α-syn operates downstream of the mitochondrial fusion/fission machinery. These novel results indicate that wild-type α-syn localizes to the MAM and modulates mitochondrial morphology, and that these behaviors are impaired by pathogenic mutations in α-syn. We believe that our results have far-reaching implications for both our understanding of α-syn biology and the treatment of synucleinopathies.


Subject(s)
Endoplasmic Reticulum/chemistry , Mitochondria/chemistry , alpha-Synuclein/analysis , Animals , Cells, Cultured , Female , HeLa Cells , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic
15.
J Neurochem ; 128(2): 330-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117942

ABSTRACT

Autosomal-dominant Alzheimer's disease (ADAD) is a genetic disorder caused by mutations in Amyloid Precursor Protein (APP) or Presenilin (PSEN) genes. Studying the mechanisms underlying these mutations can provide insight into the pathways that lead to AD pathology. The majority of biochemical studies on APP mutations to-date have focused on comparing mechanisms between mutations at different codons. It has been assumed that amino acid position is a major determinant of protein dysfunction and clinical phenotype. However, the differential effect of mutations at the same codon has not been sufficiently addressed. In the present study we compared the effects of the aggressive ADAD-associated APP I716F mutation with I716V and I716T on APP processing in human neuroglioma and CHO-K1 cells. All APP I716 mutations increased the ratio of Aß42/40 and changed the product line preference of γ-secretase towards Aß38 production. In addition, the APP I716F mutation impaired the ε-cleavage and the fourth cleavage of γ-secretase and led to abnormal APP ß-CTF accumulation at the plasma membrane. Taken together, these data indicate that APP mutations at the same codon can induce diverse abnormalities in APP processing, some resembling PSEN1 mutations. These differential effects could explain the clinical differences observed among ADAD patients bearing different APP mutations at the same position. The amyloid precursor protein (APP) I716F mutation is associated with autosomal dominant Alzheimer's disease with the youngest age-at-onset for the APP locus. Here, we describe that this mutation, when compared to two other familial Alzheimer's disease mutations at the same codon (I716V and I716T), interfered distinctly with γ-secretase cleavage. While all three mutations direct γ-secretase cleavage towards the 48→38 production line, the APP I716F mutation also impaired the ε-cleavage and the fourth cleavage of γ-secretase, resembling a PSEN1 mutation. These features may contribute to the aggressiveness of this mutation.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Adult , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Codon , Cricetinae , Cricetulus , Humans , Mutation
16.
Acta Neuropathol ; 125(2): 201-13, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23224319

ABSTRACT

Autosomal-dominant Alzheimer disease (ADAD) is a genetic disorder caused by mutations in Amyloid Precursor Protein (APP) or Presenilin (PSEN) genes. Studies from families with ADAD have been critical to support the amyloid cascade hypothesis of Alzheimer disease (AD), the basis for the current development of amyloid-based disease-modifying therapies in sporadic AD (SAD). However, whether the pathological changes in APP processing in the CNS in ADAD are similar to those observed in SAD remains unclear. In this study, we measured ß-site APP-cleaving enzyme (BACE) protein levels and activity, APP and APP C-terminal fragments in brain samples from subjects with ADAD carrying APP or PSEN1 mutations (n = 18), patients with SAD (n = 27) and age-matched controls (n = 22). We also measured sAPPß and BACE protein levels, as well as BACE activity, in CSF from individuals carrying PSEN1 mutations (10 mutation carriers and 7 non-carrier controls), patients with SAD (n = 32) and age-matched controls (n = 11). We found that in the brain, the pattern in ADAD was characterized by an increase in APP ß-C-terminal fragment (ß-CTF) levels despite no changes in BACE protein levels or activity. In contrast, the pattern in SAD in the brain was mainly characterized by an increase in BACE levels and activity, with less APP ß-CTF accumulation than ADAD. In the CSF, no differences were found between groups in BACE activity or expression or sAPPß levels. Taken together, these data suggest that the physiopathological events underlying the chronic Aß production/clearance imbalance in SAD and ADAD are different. These differences should be considered in the design of intervention trials in AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Central Nervous System/metabolism , Adult , Aged , Alzheimer Disease/cerebrospinal fluid , Amyloid Precursor Protein Secretases/cerebrospinal fluid , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/cerebrospinal fluid , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Aspartic Acid Endopeptidases/cerebrospinal fluid , Aspartic Acid Endopeptidases/metabolism , Blotting, Western , Female , Heterozygote , Humans , Immunohistochemistry , Male , Middle Aged , Mutation/genetics , Neurites/pathology , Presenilin-1/cerebrospinal fluid , Presenilin-1/genetics
17.
EMBO J ; 31(21): 4106-23, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-22892566

ABSTRACT

Alzheimer disease (AD) is associated with aberrant processing of the amyloid precursor protein (APP) by γ-secretase, via an unknown mechanism. We recently showed that presenilin-1 and -2, the catalytic components of γ-secretase, and γ-secretase activity itself, are highly enriched in a subcompartment of the endoplasmic reticulum (ER) that is physically and biochemically connected to mitochondria, called mitochondria-associated ER membranes (MAMs). We now show that MAM function and ER-mitochondrial communication-as measured by cholesteryl ester and phospholipid synthesis, respectively-are increased significantly in presenilin-mutant cells and in fibroblasts from patients with both the familial and sporadic forms of AD. We also show that MAM is an intracellular detergent-resistant lipid raft (LR)-like domain, consistent with the known presence of presenilins and γ-secretase activity in rafts. These findings may help explain not only the aberrant APP processing but also a number of other biochemical features of AD, including altered lipid metabolism and calcium homeostasis. We propose that upregulated MAM function at the ER-mitochondrial interface, and increased cross-talk between these two organelles, may play a hitherto unrecognized role in the pathogenesis of AD.


Subject(s)
Alzheimer Disease/pathology , Embryo, Mammalian/pathology , Fibroblasts/pathology , Membrane Microdomains/pathology , Mitochondria/pathology , Mitochondrial Membranes/pathology , Presenilin-1/physiology , Presenilin-2/physiology , Alzheimer Disease/metabolism , Animals , Blotting, Western , Cells, Cultured , Embryo, Mammalian/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Fibroblasts/metabolism , Humans , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Presenilin-1/antagonists & inhibitors , Presenilin-2/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions
18.
Sci Transl Med ; 4(141): 141ra90, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22764206

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder caused by genetic and environmental factors that results in degeneration of the nigrostriatal dopaminergic pathway in the brain. We analyzed neural cells generated from induced pluripotent stem cells (iPSCs) derived from PD patients and presymptomatic individuals carrying mutations in the PINK1 (PTEN-induced putative kinase 1) and LRRK2 (leucine-rich repeat kinase 2) genes, and compared them to those of healthy control subjects. We measured several aspects of mitochondrial responses in the iPSC-derived neural cells including production of reactive oxygen species, mitochondrial respiration, proton leakage, and intraneuronal movement of mitochondria. Cellular vulnerability associated with mitochondrial dysfunction in iPSC-derived neural cells from familial PD patients and at-risk individuals could be rescued with coenzyme Q(10), rapamycin, or the LRRK2 kinase inhibitor GW5074. Analysis of mitochondrial responses in iPSC-derived neural cells from PD patients carrying different mutations provides insight into convergence of cellular disease mechanisms between different familial forms of PD and highlights the importance of oxidative stress and mitochondrial dysfunction in this neurodegenerative disease.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Mitochondria/drug effects , Mitochondria/pathology , Neurons/cytology , Neurons/metabolism , Parkinson Disease/metabolism , Humans , Indoles/therapeutic use , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Neurons/drug effects , Phenols/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sirolimus/therapeutic use , Ubiquinone/therapeutic use
19.
Mov Disord ; 27(3): 343, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22512003
20.
EMBO Mol Med ; 4(7): 660-73, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22488900

ABSTRACT

Inherited familial Alzheimer's disease (AD) is characterized by small increases in the ratio of Aß42 versus Aß40 peptide which is thought to drive the amyloid plaque formation in the brain of these patients. Little is known however whether ageing, the major risk factor for sporadic AD, affects amyloid beta-peptide (Aß) generation as well. Here we demonstrate that the secretion of Aß is enhanced in an in vitro model of neuronal ageing, correlating with an increase in γ-secretase complex formation. Moreover we found that peroxynitrite (ONOO(-)), produced by the reaction of superoxide anion with nitric oxide, promoted the nitrotyrosination of presenilin 1 (PS1), the catalytic subunit of γ-secretase. This was associated with an increased association of the two PS1 fragments, PS1-CTF and PS1-NTF, which constitute the active catalytic centre. Furthermore, we found that peroxynitrite shifted the production of Aß towards Aß(42) and increased the Aß(42) /Aß(40) ratio. Our work identifies nitrosative stress as a potential mechanistic link between ageing and AD.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/metabolism , Cellular Senescence/drug effects , Neurons/metabolism , Peroxynitrous Acid/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Catalytic Domain , Cells, Cultured , Humans , Mice , Neurons/cytology , Peptide Fragments/metabolism , Presenilin-1/chemistry , Presenilin-1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Risk Factors , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...