Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Scand J Work Environ Health ; 48(1): 5-20, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34582561

ABSTRACT

OBJECTIVES: This systematic review and meta-analysis aimed to synthesize the available data on prospective associations between work-related stressors and the risk of type 2 diabetes mellitus (T2DM) among adult workers, according to the demand-control-support (DCS) and the effort-reward imbalance (ERI) models. METHOD: We searched for prospective studies in PubMed, EMBASE, Web of Science, Scopus, CINHAL and PsychInfo. After screening and extraction, quality of evidence was assessed using the ROBINS-I tool adapted for observational studies. The effect estimates extracted for each cohort were synthesized using random effect models. RESULTS: We included 18 studies (reporting data on 25 cohorts) in meta-analyses for job strain, job demands, job control, social support at work and ERI. Workers exposed to job strain had a higher risk of developing T2DM when compared to unexposed workers [pooled rate ratio (RR) 1.16, 95% confidence interval (CI) 1.07-1.26]. This association was robust in several supplementary analyses. For exposed women relative to unexposed women, the RR was 1.35 (95% CI 1.12-1.64). The RR of workers exposed to ERI was 1.24 (95% CI 1.08-1.42) compared to unexposed workers. CONCLUSIONS: This is the first meta-analysis to find an effect of ERI on the onset of T2DM incidence. It also confirms that job strain increases the incidence of T2DM, especially among women.


Subject(s)
Diabetes Mellitus, Type 2 , Adult , Diabetes Mellitus, Type 2/epidemiology , Female , Humans , Prospective Studies , Reward , Risk Factors , Stress, Psychological/complications , Stress, Psychological/epidemiology
2.
J Neurochem ; 156(1): 88-105, 2021 01.
Article in English | MEDLINE | ID: mdl-31886886

ABSTRACT

Insulin signaling through the insulin receptor has long been studied in classic target organs, such as adipose tissue and skeletal muscle, where one of its effects is to increase glucose uptake. Insulin and insulin receptor are present in many areas of the brain, but the functions of brain insulin signaling outside feeding circuits are not well defined. It has been proposed that hippocampal insulin signaling is important for memory, that brain insulin signaling is deficient in Alzheimer's disease, and that intranasal insulin treatment improves cognition, but the mechanisms remain unclear and do not seem to involve increased glucose uptake by neurons. The molecular behavior of the insulin receptor itself is not well known in living neurons; therefore, we investigated the spatial dynamics of the insulin receptor on somatodendritic membranes of live rat hippocampal neurons in culture. Using single-molecule tracking of quantum dot-tagged insulin receptors and single-particle tracking photoactivation localization microscopy, we show that the insulin receptor is distributed over both dendritic shafts and spines. Using colocalization with synaptic markers, we also show that in contrast to the glutamate receptor subunit glutamate receptor subunit A1, the dynamics of the insulin receptor are not affected by association with excitatory synapses; however, the insulin receptor is immobilized by components of inhibitory synapses. The mobility of the insulin receptor is reduced both by low concentrations of the pro-inflammatory cytokine tumor necrosis factor α and by cholesterol depletion, suggesting an association with sphingolipid-rich membrane domains. On the other hand, the insulin receptor dynamics in hippocampal neurons are not affected by increased excitatory signaling. Finally, using real-time single-event quantification, we find evidence of strong insulin receptor exocytosis on dendritic shafts. Our results suggest an association of the neuronal insulin receptor with specific elements of the dendritic shaft, rather than excitatory synapses.


Subject(s)
Dendrites/metabolism , Hippocampus/metabolism , Receptor, Insulin/metabolism , Animals , Cells, Cultured , Female , Male , Neurons/metabolism , Rats , Rats, Sprague-Dawley
3.
J Pathol ; 245(1): 85-100, 2018 05.
Article in English | MEDLINE | ID: mdl-29435980

ABSTRACT

Alzheimer's disease (AD) is a devastating neurological disorder that still lacks an effective treatment, and this has stimulated an intense pursuit of disease-modifying therapeutics. Given the increasingly recognized link between AD and defective brain insulin signaling, we investigated the actions of liraglutide, a glucagon-like peptide-1 (GLP-1) analog marketed for treatment of type 2 diabetes, in experimental models of AD. Insulin receptor pathology is an important feature of AD brains that impairs the neuroprotective actions of central insulin signaling. Here, we show that liraglutide prevented the loss of brain insulin receptors and synapses, and reversed memory impairment induced by AD-linked amyloid-ß oligomers (AßOs) in mice. Using hippocampal neuronal cultures, we determined that the mechanism of neuroprotection by liraglutide involves activation of the PKA signaling pathway. Infusion of AßOs into the lateral cerebral ventricle of non-human primates (NHPs) led to marked loss of insulin receptors and synapses in brain regions related to memory. Systemic treatment of NHPs with liraglutide provided partial protection, decreasing AD-related insulin receptor, synaptic, and tau pathology in specific brain regions. Synapse damage and elimination are amongst the earliest known pathological changes and the best correlates of memory impairment in AD. The results illuminate mechanisms of neuroprotection by liraglutide, and indicate that GLP-1 receptor activation may be harnessed to protect brain insulin receptors and synapses in AD. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cognitive Dysfunction/drug therapy , Liraglutide/pharmacology , Memory/drug effects , Receptor, Insulin/drug effects , Synapses/pathology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Hippocampus/drug effects , Hypoglycemic Agents/pharmacology , Male , Mice , Receptor, Insulin/metabolism , Synapses/drug effects
4.
Biochim Biophys Acta Gen Subj ; 1862(4): 1050-1068, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29292190

ABSTRACT

Relationship of donor and acceptor fluorescence anisotropies as well as efficiency of fluorescence resonance energy transfer (FRET) has been investigated in a confocal microscope in the context of FRET systems comprised of donor and acceptor-labeled MHCI and MHCII receptors on the surface of Kit-225 K6 human T-cells. The measurements have been carried out in a 2-laser, 5-signal platform where the total donor fluorescence intensity and 2 acceptor fluorescence intensities with their anisotropies - one at the donor's excitation wavelength, the other at the acceptor's excitation wavelength - have been detected. This configuration enabled the determination of FRET efficiency and correlating it with the two acceptor fluorescence anisotropies as a kind of calibration. Estimations for the FRET-enhanced donor fluorescence anisotropy, the directly excited acceptor fluorescence anisotropy, and the fluorescence anisotropy of sensitized emission have been obtained. Procedures for determining FRET by measuring only the total donor intensity and the acceptor intensity and its anisotropy, or two acceptor intensities and their anisotropies have been elaborated, the errors of which have been estimated based on the fluorescence anisotropy values obtained in the calibration with the method of flow cytometric energy transfer (FCET). The combined detection of the donor and acceptor fluorescence anisotropies enabled also the determination of the lower and upper limits of the orientation factor for FRET (κ2). An increase in range for κ2 with increasing FRET efficiency has been observed, with average κ2 values different from the dynamic random average of 2/3. These observations call for the need of κ2 determination in proximity measurements, where the donor and acceptor orientations are not predictable. An increasing range of κ2 with increasing intermolecular proximity of the MHCI and MHCII receptors has been observed. This indicates that molecular flexibility in the clusters of the MHCI and MHCII receptors reduces with increasing cluster density, i.e. a "fluidity gradient" exists in the clusters. More specifically, the local density dependent flexibility can also be taken as a direct proof for that the association of these receptors is non-random, but mediated by some type of physical interaction, a finding as a benefit of FRET detection by polarization spectroscopy. Two new quantities - the quenched donor fluorescence anisotropy and a fluorescence anisotropy analogue, the "dissymmetry index" of the polarized FRET efficiency components - have also been introduced for the characterization of the orientational dynamics of the excited state during FRET.


Subject(s)
Cell Membrane/metabolism , Fluorescence Polarization/methods , Fluorescence Resonance Energy Transfer/methods , Microscopy, Confocal/methods , Algorithms , Anisotropy , Cell Line, Tumor , Cell Membrane/chemistry , Flow Cytometry/methods , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/metabolism , Humans , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/metabolism
5.
Behav Brain Res ; 333: 150-160, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28668282

ABSTRACT

Parkinson's disease (PD) is characterized by motor dysfunction, which is preceded by a number of non-motor symptoms including olfactory deficits. Aggregation of α-synuclein (α-syn) gives rise to Lewy bodies in dopaminergic neurons and is thought to play a central role in PD pathology. However, whether amyloid fibrils or soluble oligomers of α-syn are the main neurotoxic species in PD remains controversial. Here, we performed a single intracerebroventricular (i.c.v.) infusion of α-syn oligomers (α-SYOs) in mice and evaluated motor and non-motor symptoms. Familiar bedding and vanillin essence discrimination tasks showed that α-SYOs impaired olfactory performance of mice, and decreased TH and dopamine levels in the olfactory bulb early after infusion. The olfactory deficit persisted until 45days post-infusion (dpi). α- SYO-infused mice behaved normally in the object recognition and forced swim tests, but showed increased anxiety-like behavior in the open field and elevated plus maze tests 20 dpi. Finally, administration of α-SYOs induced late motor impairment in the pole test and rotarod paradigms, along with reduced TH and dopamine content in the caudate putamen, 45 dpi. Reduced number of TH-positive cells was also seen in the substantia nigra of α-SYO-injected mice compared to control. In conclusion, i.c.v. infusion of α-SYOs recapitulated some of PD-associated non-motor symptoms, such as increased anxiety and olfactory dysfunction, but failed to recapitulate memory impairment and depressive-like behavior typical of the disease. Moreover, α-SYOs i.c.v. administration induced motor deficits and loss of TH and dopamine levels, key features of PD. Results point to α-syn oligomers as the proximal neurotoxins responsible for early non-motor and motor deficits in PD and suggest that the i.c.v. infusion model characterized here may comprise a useful tool for identification of PD novel therapeutic targets and drug screening.


Subject(s)
Behavioral Symptoms/etiology , Brain/drug effects , Olfaction Disorders/etiology , Parkinson Disease/complications , Parkinson Disease/etiology , alpha-Synuclein/toxicity , Animals , Brain/metabolism , Cells, Cultured , Discrimination, Psychological/drug effects , Disease Models, Animal , Embryo, Mammalian , Humans , Injections, Intraventricular , Male , Maze Learning/drug effects , Mesencephalon/cytology , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Peptides/toxicity , Recognition, Psychology/drug effects , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
6.
J Neurochem ; 140(3): 359-367, 2017 02.
Article in English | MEDLINE | ID: mdl-27889917

ABSTRACT

Insulin is known mainly for its effects in peripheral tissues, such as the liver, skeletal muscles and adipose tissue, where the activation of the insulin receptor (IR) has both short-term and long-term effects. Insulin and the IR are also present in the brain, and since there is evidence that neuronal insulin signaling regulates synaptic plasticity and that it is impaired in disease, this pathway might be the key to protection or reversal of symptoms, especially in Alzheimer's disease. However, there are controversies about the importance of the neuronal IR, partly because biophysical data on its activation and signaling are much less complete than for the peripheral IR. This review briefly summarizes the neuronal IR signaling in health and disease, and then focuses on known differences between the neuronal and peripheral IR with regard to alternative splicing and glycosylation, and lack of data with respect to phosphorylation and membrane subdomain localization. Particularities in the neuronal IR itself and its environment may have consequences for downstream signaling and impact synaptic plasticity. Furthermore, establishing the relative importance of insulin signaling through IR or through hybrids with its homolog, the insulin-like growth factor 1 receptor, is crucial for evaluating the consequences of brain IR activation. An improved biophysical understanding of the neuronal IR may help predict the consequences of insulin-targeted interventions.


Subject(s)
Antigens, CD/metabolism , Brain/metabolism , Neurons/metabolism , Receptor, Insulin/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Brain/drug effects , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin/therapeutic use , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/drug effects , Receptor, Insulin/agonists
7.
BMC Evol Biol ; 16: 75, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068704

ABSTRACT

BACKGROUND: Brain-expressed proteins that have undergone functional change during human evolution may contribute to human cognitive capacities, and may also leave us vulnerable to specifically human diseases, such as schizophrenia, autism or Alzheimer's disease. In order to search systematically for those proteins that have changed the most during human evolution and that might contribute to brain function and pathology, all proteins with orthologs in chimpanzee, orangutan and rhesus macaque and annotated as being expressed on the surface of cells in the human central nervous system were ordered by the number of human-specific amino acid differences that are fixed in modern populations. RESULTS: PCDHB11, a beta-protocadherin homologous to murine cell adhesion proteins, stood out with 12 substitutions and maintained its lead after normalizing for protein size and applying weights for amino acid exchange probabilities. Human PCDHB11 was found to cause homophilic cell adhesion, but at lower levels than shown for other clustered protocadherins. Homophilic adhesion caused by a PCDHB11 with reversion of human-specific changes was as low as for modern human PCDHB11; while neither human nor reverted PCDHB11 adhered to controls, they did adhere to each other. A loss of function in PCDHB11 is unlikely because intra-human variability did not increase relative to the other human beta-protocadherins. CONCLUSIONS: The brain-expressed protein with the highest number of human-specific substitutions is PCDHB11. In spite of its fast evolution and low intra-human variability, cell-based tests on the only proposed function for PCDHB11 did not indicate a functional change.


Subject(s)
Brain/physiology , Cadherins/genetics , Evolution, Molecular , Nerve Tissue Proteins/genetics , Animals , Biological Evolution , Brain/pathology , Cadherins/metabolism , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cytological Techniques , Humans , K562 Cells , Macaca mulatta/genetics , Mice , Nerve Tissue Proteins/metabolism , Pan troglodytes/genetics , Pongo abelii/genetics , Protocadherins
8.
J Neurosci ; 34(41): 13629-43, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25297091

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disorder and a major medical problem. Here, we have investigated the impact of amyloid-ß (Aß) oligomers, AD-related neurotoxins, in the brains of rats and adult nonhuman primates (cynomolgus macaques). Soluble Aß oligomers are known to accumulate in the brains of AD patients and correlate with disease-associated cognitive dysfunction. When injected into the lateral ventricle of rats and macaques, Aß oligomers diffused into the brain and accumulated in several regions associated with memory and cognitive functions. Cardinal features of AD pathology, including synapse loss, tau hyperphosphorylation, astrocyte and microglial activation, were observed in regions of the macaque brain where Aß oligomers were abundantly detected. Most importantly, oligomer injections induced AD-type neurofibrillary tangle formation in the macaque brain. These outcomes were specifically associated with Aß oligomers, as fibrillar amyloid deposits were not detected in oligomer-injected brains. Human and macaque brains share significant similarities in terms of overall architecture and functional networks. Thus, generation of a macaque model of AD that links Aß oligomers to tau and synaptic pathology has the potential to greatly advance our understanding of mechanisms centrally implicated in AD pathogenesis. Furthermore, development of disease-modifying therapeutics for AD has been hampered by the difficulty in translating therapies that work in rodents to humans. This new approach may be a highly relevant nonhuman primate model for testing therapeutic interventions for AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Peptide Fragments/toxicity , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/administration & dosage , Animals , Apoptosis/drug effects , Astrocytes/pathology , Injections, Intraventricular , Macaca fascicularis , Male , Microglia/pathology , Microinjections , Neurofibrillary Tangles/pathology , Peptide Fragments/administration & dosage , Rats , Rats, Wistar , Synapses/pathology , Synapses/physiology , Synapses/ultrastructure
9.
PLoS One ; 6(7): e22857, 2011.
Article in English | MEDLINE | ID: mdl-21829538

ABSTRACT

The amyloid precursor protein (APP) is well known for giving rise to the amyloid-ß peptide and for its role in Alzheimer's disease. Much less is known, however, on the physiological roles of APP in the development and plasticity of the central nervous system. We have used phage display of a peptide library to identify high-affinity ligands of purified recombinant human sAPPα(695) (the soluble, secreted ectodomain from the main neuronal APP isoform). Two peptides thus selected exhibited significant homologies with the conserved extracellular domain of several members of the semaphorin (Sema) family of axon guidance proteins. We show that sAPPα(695) binds both purified recombinant Sema3A and Sema3A secreted by transfected HEK293 cells. Interestingly, sAPPα(695) inhibited the collapse of embryonic chicken (Gallus gallus domesticus) dorsal root ganglia growth cones promoted by Sema3A (K(d)≤8·10(-9) M). Two Sema3A-derived peptides homologous to the peptides isolated by phage display blocked sAPPα binding and its inhibitory action on Sema3A function. These two peptides are comprised within a domain previously shown to be involved in binding of Sema3A to its cellular receptor, suggesting a competitive mechanism by which sAPPα modulates the biological action of semaphorins.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Ganglia, Spinal/metabolism , Growth Cones/physiology , Peptide Fragments/metabolism , Semaphorin-3A/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , Chick Embryo , Culture Media, Conditioned/pharmacology , Ganglia, Spinal/cytology , Humans , Immunoprecipitation , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Peptide Library , Protein Conformation
10.
Mol Biol Evol ; 28(1): 25-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20798139

ABSTRACT

With the sequencing of the Neandertal genome, it has become possible to identify amino acid substitutions that occurred on the human lineage since its separation from the Neandertal lineage. Conceptually, it will therefore be possible to functionally analyze all such amino acid substitutions in the future. Here, we analyze the function of substitutions that occurred during recent human evolution in N-terminal signal peptides. We develop a high-throughput flow cytometry-based assay to analyze signal peptide efficiency as the ratio of surface to total reporter protein per live cell. Such ratios differed significantly among signal peptides derived from different human genes. However, no modern human signal peptide differed significantly from its ancestral counterpart, an observation compatible with the predictions of the neutral theory of molecular evolution.


Subject(s)
Amino Acid Substitution , Fossils , Genome, Human , Protein Sorting Signals/genetics , Sequence Analysis, DNA , Amino Acid Sequence , Evolution, Molecular , Humans , Molecular Sequence Data
11.
Biochem Biophys Res Commun ; 402(4): 747-9, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20977882

ABSTRACT

Targeting of nuclear-encoded proteins to different organelles, such as mitochondria, is a process that can result in the redeployment of proteins to new intracellular destinations during evolution. With the sequencing of the Neandertal genome, it has become possible to identify amino acid substitutions that occurred on the modern human lineage since its separation from the Neandertal lineage. Here we analyze the function of two substitutions in mitochondrial targeting sequences that occurred and rose to high frequency recently during recent human evolution. The ancestral and modern versions of the two targeting sequences do not differ in the efficiency with which they direct a protein to the mitochondria, an observation compatible with the neutral theory of molecular evolution.


Subject(s)
Amino Acid Substitution/genetics , DNA, Mitochondrial/genetics , Evolution, Molecular , Genetic Speciation , Genome, Human/genetics , Base Sequence , Cell Line , Genetic Testing , Humans , Phylogeny
12.
J Biol Chem ; 284(22): 15016-25, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19336403

ABSTRACT

The amyloid precursor protein (APP) is implied both in cell growth and differentiation and in neurodegenerative processes in Alzheimer disease. Regulated proteolysis of APP generates biologically active fragments such as the neuroprotective secreted ectodomain sAPPalpha and the neurotoxic beta-amyloid peptide. Furthermore, it has been suggested that the intact transmembrane APP plays a signaling role, which might be important for both normal synaptic plasticity and neuronal dysfunction in dementia. To understand APP signaling, we tracked single molecules of APP using quantum dots and quantitated APP homodimerization using fluorescence lifetime imaging microscopy for the detection of Förster resonance energy transfer in living neuroblastoma cells. Using selective labeling with synthetic fluorophores, we show that the dimerization of APP is considerably higher at the plasma membrane than in intracellular membranes. Heparan sulfate significantly contributes to the almost complete dimerization of APP at the plasma membrane. Importantly, this technique for the first time structurally defines the initiation of APP signaling by binding of a relevant physiological extracellular ligand; our results indicate APP as receptor for neuroprotective sAPPalpha, as sAPPalpha binding disrupts APP dimers, and this disruption of APP dimers by sAPPalpha is necessary for the protection of neuroblastoma cells against starvation-induced cell death. Only cells expressing reversibly dimerized wild-type, but not covalently dimerized mutant APP are protected by sAPPalpha. These findings suggest a potentially beneficial effect of increasing sAPPalpha production or disrupting APP dimers for neuronal survival.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Neuroprotective Agents/metabolism , Protein Multimerization , Acyl Carrier Protein , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival , Green Fluorescent Proteins/metabolism , Heparin/metabolism , Heparin/pharmacology , Models, Biological , Neuroblastoma/pathology , Protein Multimerization/drug effects , Quantum Dots
13.
Biochemistry ; 47(49): 13115-26, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-19007185

ABSTRACT

Intracellular pH is an important indicator for cellular metabolism and pathogenesis. pH sensing in living cells has been achieved using a number of synthetic organic dyes and genetically expressible sensor proteins, even allowing the specific targeting of intracellular organelles. Ideally, a class of genetically encodeable sensors need to cover relevant cellular pH ranges. We present a FRET-based pH sensor platform, based on the pH modulation of YFP acceptor fluorophores in a fusion construct with ECFP. The concurrent loss of the overlap integral upon acidification results in a proportionally reduced FRET coupling. The readout of FRET over the sensitized YFP fluorescence lifetime yields a highly sensitive and robust pH measurement that is self-calibrated. The principle is demonstrated in the existing high-efficiency FRET fusion Cy11.5, and tunability of the platform design is demonstrated by genetic alteration of the pH sensitivity of the acceptor moiety.


Subject(s)
Biotechnology/methods , Chemistry Techniques, Analytical/methods , Fluorescence Resonance Energy Transfer/methods , Luminescent Proteins/chemistry , Animals , Cell Line , Cells, Cultured , Hydrogen-Ion Concentration , Image Processing, Computer-Assisted , Luminescent Proteins/analysis , Sensitivity and Specificity
14.
J Neurosci ; 28(11): 2874-82, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18337418

ABSTRACT

The flotillins/reggie proteins are associated with noncaveolar membrane microdomains and have been implicated in the regulation of a clathrin- and caveolin-independent endocytosis pathway. Endocytosis is required for the amyloidogenic processing of the amyloid precursor protein (APP) and thus to initiate the release of the neurotoxic beta-amyloid peptide (Abeta), the major component of extracellular plaques found in the brains of Alzheimer's disease patients. Here, we report that small interference RNA-mediated downregulation of flotillin-2 impairs the endocytosis of APP, in both neuroblastoma cells and primary cultures of hippocampal neurons, and reduces the production of Abeta. Similar to tetanus neurotoxin endocytosis, but unlike the internalization of transferrin, clathrin-dependent endocytosis of APP requires cholesterol and adaptor protein-2 but is independent of epsin1 function. Moreover, on a nanoscale resolution using stimulated emission depletion microscopy and by Förster resonance energy transfer with fluorescence lifetime imaging microscopy, we provide evidence that flotillin-2 promotes the clustering of APP at the cell surface. We show that the interaction of flotillin-2 with APP is dependent on cholesterol and that clustering of APP enhances its endocytosis rate. Together, our data suggest that cholesterol/flotillin-dependent clustering of APP may stimulate the internalization into a specialized clathrin-dependent endocytosis pathway to promote amyloidogenic processing.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Endocytosis/physiology , Membrane Proteins/metabolism , Neurons/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Membrane Proteins/genetics , Mice , Mice, Transgenic
15.
Prog Neurobiol ; 82(1): 11-32, 2007 May.
Article in English | MEDLINE | ID: mdl-17428603

ABSTRACT

The amyloid precursor protein (APP) is a transmembrane protein that plays major roles in the regulation of several important cellular functions, especially in the nervous system, where it is involved in synaptogenesis and synaptic plasticity. The secreted extracellular domain of APP, sAPPalpha, acts as a growth factor for many types of cells and promotes neuritogenesis in post-mitotic neurons. Alternative proteolytic processing of APP releases potentially neurotoxic species, including the amyloid-beta (Abeta) peptide that is centrally implicated in the pathogenesis of Alzheimer's disease (AD). Reinforcing this biochemical link to neuronal dysfunction and neurodegeneration, APP is also genetically linked to AD. In this review, we discuss the biological functions of APP in the context of tissue morphogenesis and restructuring, where APP appears to play significant roles both as a contact receptor and as a diffusible factor. Structural investigation of APP, which is necessary for a deeper understanding of its roles at a molecular level, has also been advancing rapidly. We summarize recent progress in the determination of the structure of isolated APP fragments and of the conformations of full-length sAPPalpha, in both monomeric and dimeric states. The potential role of APP dimerization for the regulation of its biological functions is also discussed.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/physiology , Brain/physiology , Amyloid beta-Protein Precursor/genetics , Animals , Humans , Protein Structure, Tertiary
16.
J Mol Biol ; 357(2): 493-508, 2006 Mar 24.
Article in English | MEDLINE | ID: mdl-16436282

ABSTRACT

Proteolytic cleavage of the amyloid precursor protein (APP) by beta and gamma-secretases gives rise to the beta-amyloid peptide, considered to be a causal factor in Alzheimer's disease. Conversely, the soluble extracellular domain of APP (sAPPalpha), released upon its cleavage by alpha-secretase, plays a number of important physiological functions. Several APP fragments have been structurally characterized at atomic resolution, but the structures of intact APP and of full-length sAPPalpha have not been determined. Here, ab initio reconstruction of molecular models from high-resolution solution X-ray scattering (SAXS) data for the two main isoforms of sAPPalpha (sAPPalpha(695) and sAPPalpha(770)) provided models of sufficiently high resolution to identify distinct structural domains of APP. The fragments for which structures are known at atomic resolution were fitted within the solution models of full-length sAPPalpha, allowing localization of important functional sites (i.e. glycosylation, protease inhibitory and heparin-binding sites). Furthermore, combined results from SAXS, analytical ultracentrifugation (AUC) and size-exclusion chromatography (SEC) analysis indicate that both sAPPalpha isoforms are monomeric in solution. On the other hand, SEC, bis-ANS fluorescence, AUC and SAXS measurements showed that sAPPalpha forms a 2:1 complex with heparin. A conformational model for the sAPPalpha:heparin complex was also derived from the SAXS data. Possible implications of such complex formation for the physiological dimerization of APP and biological signaling are discussed in terms of the structural models proposed.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Heparin/metabolism , Protein Isoforms/chemistry , Protein Structure, Quaternary , Protein Structure, Tertiary , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cell Membrane/chemistry , Dimerization , Heparin/chemistry , Humans , Models, Molecular , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Ultracentrifugation
17.
J Biol Chem ; 278(36): 34259-67, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12796495

ABSTRACT

The beta-amyloid peptide (A beta), the major component of the senile plaques found in the brains of Alzheimer's disease patients, is derived from proteolytic processing of a transmembrane glycoprotein known as the amyloid precursor protein (APP). Human APP exists in various isoforms, of which the major ones contain 695, 751, and 770 amino acids. Proteolytic cleavage of APP by alpha- or beta-secretases releases the extracellular soluble fragments sAPP alpha or sAPP beta, respectively. Despite the fact that sAPP alpha plays important roles in both physiological and pathological processes in the brain, very little is known about its structure and stability. We have recently presented a structural model of sAPP alpha 695 obtained from small-angle x-ray scattering measurements (Gralle, M., Botelho, M. M., Oliveira, C. L. P., Torriani, I., and Ferreira, S. T. (2002) Biophys. J. 83, 3513-3524). We now report studies on the folding and stabilities of sAPP alpha 695 and sAPP alpha 770. The combined use of intrinsic fluorescence, 4-4'-Dianilino-1,1'binaphthyl-5,5'-disulfonic acid (bis-ANS) fluorescence, circular dichroism, differential ultraviolet absorption, and small-angle x-ray scattering measurements of the equilibrium unfolding of sAPP alpha 695 and sAPP alpha 770 by GdnHCl and urea revealed multistep folding pathways for both sAPP alpha isoforms. Such stepwise folding processes may be related to the identification of distinct structural domains in the three-dimensional model of sAPP alpha. Furthermore, the relatively low stability of the native state of sAPP alpha suggests that conformational plasticity may play a role in allowing APP to interact with a number of distinct physiological ligands.


Subject(s)
Amyloid/chemistry , Anilino Naphthalenesulfonates/pharmacology , Circular Dichroism , Dose-Response Relationship, Drug , Guanidine/pharmacology , Humans , Kinetics , Ligands , Microscopy, Fluorescence , Protein Conformation , Protein Folding , Protein Isoforms , Protein Structure, Tertiary , Scattering, Radiation , Ultraviolet Rays , Urea/pharmacology , X-Rays
18.
Biophys J ; 83(6): 3513-24, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12496118

ABSTRACT

The amyloid precursor protein (APP) is the precursor of the beta-amyloid peptide (Abeta), which is centrally related to the genesis of Alzheimer's disease (AD). In addition, APP has been suggested to mediate and/or participate in events that lead to neuronal degeneration in AD. Despite the fact that various aspects of the cell biology of APP have been investigated, little information on the structure of this protein is available. In this work, the solution structure of the soluble extracellular domain of APP (sAPP, composing 89% of the amino acid residues of the whole protein) has been investigated through a combination of size-exclusion chromatography, circular dichroism, and synchrotron radiation small-angle x-ray scattering (SAXS) studies. sAPP is monomeric in solution (65 kDa obtained from SAXS measurements) and exhibits an anisometric molecular shape, with a Stokes radius of 39 or 51 A calculated from SAXS or chromatographic data, respectively. The radius of gyration and the maximum molecular length obtained by SAXS were 38 A and 130 A, respectively. Analysis of SAXS data further allowed building a structural model for sAPP in solution. Circular dichroism data and secondary structure predictions based on the amino acid sequence of APP suggested that a significant fraction of APP (30% of the amino acid residues) is not involved in standard secondary structure elements, which may explain the elongated shape of the molecule recovered in our structural model. Possible implications of the structure of APP in ligand binding and molecular recognition events involved in the biological functions of this protein are discussed.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Solutions/chemistry , Chromatography, Gel/methods , Circular Dichroism/methods , Computer Simulation , Extracellular Space/chemistry , Humans , Models, Molecular , Pichia/chemistry , Pichia/classification , Pichia/metabolism , Protein Conformation , Protein Isoforms/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Species Specificity , Water/chemistry , X-Ray Diffraction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...