Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7: 44507, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28291226

ABSTRACT

Aß peptide that accumulates in Alzheimer's disease brain, derives from proteolytic processing of the amyloid precursor protein (APP) that exists in three main isoforms derived by alternative splicing. The isoform APP695, lacking exons 7 and 8, is predominately expressed in neurons and abnormal neuronal splicing of APP has been observed in the brain of patients with Alzheimer's disease. Herein, we demonstrate that expression of the neuronal members of the ELAVL protein family (nELAVLs) correlate with APP695 levels in vitro and in vivo. Moreover, we provide evidence that nELAVLs regulate the production of APP695; by using a series of reporters we show that concurrent binding of nELAVLs to sequences located both upstream and downstream of exon 7 is required for its skipping, whereas nELAVL-binding to a highly conserved U-rich sequence upstream of exon 8, is sufficient for its exclusion. Finally, we report that nELAVLs block APP exon 7 or 8 definition by reducing the binding of the essential splicing factor U2AF65, an effect facilitated by the concurrent binding of AUF-1. Our study provides new insights into the regulation of APP pre-mRNA processing, supports the role for nELAVLs as neuron-specific splicing regulators and reveals a novel function of AUF1 in alternative splicing.


Subject(s)
Alternative Splicing/genetics , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Peptide Fragments/genetics , Alzheimer Disease/pathology , Brain/pathology , ELAV-Like Protein 2/genetics , Gene Expression Regulation , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Humans , Multigene Family/genetics , Neurons/metabolism , Neurons/pathology , Protein Binding , Protein Isoforms/genetics , RNA Precursors/genetics , Splicing Factor U2AF/genetics , T-Cell Intracellular Antigen-1/genetics
2.
Cell Rep ; 9(5): 1742-1755, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25466251

ABSTRACT

Fragile X syndrome (FXS) is the leading genetic cause of autism. Mutations in Fmr1 (fragile X mental retardation 1 gene) engender exaggerated translation resulting in dendritic spine dysmorphogenesis, synaptic plasticity alterations, and behavioral deficits in mice, which are reminiscent of FXS phenotypes. Using postmortem brains from FXS patients and Fmr1 knockout mice (Fmr1(-/y)), we show that phosphorylation of the mRNA 5' cap binding protein, eukaryotic initiation factor 4E (eIF4E), is elevated concomitant with increased expression of matrix metalloproteinase 9 (MMP-9) protein. Genetic or pharmacological reduction of eIF4E phosphorylation rescued core behavioral deficits, synaptic plasticity alterations, and dendritic spine morphology defects via reducing exaggerated translation of Mmp9 mRNA in Fmr1(-/y) mice, whereas MMP-9 overexpression produced several FXS-like phenotypes. These results uncover a mechanism of regulation of synaptic function by translational control of Mmp-9 in FXS, which opens the possibility of new treatment avenues for the diverse neurological and psychiatric aspects of FXS.


Subject(s)
Benzofurans/pharmacology , Eukaryotic Initiation Factor-4E/physiology , Fragile X Syndrome/drug therapy , Matrix Metalloproteinase 9/genetics , Protein Biosynthesis/drug effects , Adenosine Triphosphatases/antagonists & inhibitors , Animals , Autistic Disorder/enzymology , Benzofurans/therapeutic use , Brain/enzymology , Cation Transport Proteins/antagonists & inhibitors , Cells, Cultured , Copper-Transporting ATPases , Dendritic Spines/pathology , Enzyme Induction/drug effects , Female , Fragile X Syndrome/enzymology , Fragile X Syndrome/genetics , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism
3.
Neurobiol Dis ; 70: 179-89, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25008761

ABSTRACT

Accumulation of amyloid-ß (Αß) peptide is believed to play a central role in the pathogenesis of Alzheimer's disease (AD). Lowering Aß levels in the brain may thus improve synaptic and cognitive deficits observed in AD patients. In the non-amyloidogenic pathway, the amyloid-ß precursor protein (APP) is cleaved within the Aß peptide sequence by α-secretases, giving rise to the potent neurotrophic N-terminal fragment sΑPPα. We have previously reported that gelatinase B/matrix metalloproteinase 9 (MMP-9), a matrix metalloproteinase critically involved in neuronal plasticity, acts as α-secretase both in vitro and in vivo and reduces Aß levels in vitro. In the present study, we demonstrate that neuronal overexpression of MMP-9 in a transgenic AD mouse model harboring five familial AD-related mutations (5xFAD) resulted in increased sAPPα levels and decreased Aß oligomers without affecting amyloid plaque load in the brain. Functionally, overexpression of MMP-9 prevented the cognitive deficits displayed by 5xFAD mice, an improvement that was accompanied by increased levels of the pre-synaptic protein synaptophysin and mature brain-derived neurotrophic factor (BDNF) in the brain. These results suggest that in vivo activation of endogenous MMP-9 could be a promising target for interference with development and/or progression of AD.


Subject(s)
Alzheimer Disease/physiopathology , Brain/physiopathology , Matrix Metalloproteinase 9/metabolism , Neurons/physiology , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/pathology , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Female , Humans , Male , Matrix Metalloproteinase 9/genetics , Maze Learning/physiology , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Recognition, Psychology/physiology , Sex Characteristics , Synaptophysin/metabolism
4.
Front Cell Neurosci ; 8: 182, 2014.
Article in English | MEDLINE | ID: mdl-25071443

ABSTRACT

Differential expression of microRNAs (miRs) in the brain of patients with neurodegenerative diseases suggests that they may have key regulatory roles in the development of these disorders. Two such miRs, miR-7, and miR-153 have recently been shown to target α-synuclein, a protein critically involved in the pathological process of Parkinson's disease. By using a well-established in culture Parkinson's disease model that of neurotoxin 1-Methyl-4-Phenyl-Pyridinium (MPP(+)), we examined whether miR-7 and miR-153 display neuroprotective properties. Herein, we demonstrate that treatment of cortical neurons with MPP(+) induced a dose-dependent cell death with apoptotic characteristics. This was reflected in altered intracellular signaling characterized by increased levels of activated kinases p38MAPK and ERK1/2 and reduced levels of activated AKT, p70S6K, and SAPK/JNK. Overexpression of miR-7 or miR-153 by adenoviral transduction protected cortical neurons from MPP(+)-induced toxicity, restored neuronal viability and anti-apoptotic BCL-2 protein levels while attenuated activation of caspase-3. Moreover, both miR-7 and miR-153 interfered with MPP(+)-induced alterations in intracellular signaling pathways in a partially overlapping manner; specifically, they preserved activation of mTOR and SAPK/JNK signaling pathways in the MPP(+)-treated neurons, while miR-153 also attenuated MPP(+)-induced activation of p38MAPK. No major effects were observed in the rest of signaling cascades or proteins investigated. Furthermore, the neuroprotective effect of miR-7 and miR-153 was alleviated when MPP(+) was co-administered with rapamycin. Taken together, our results suggest that miR-7 and miR-153 protect neurons from cell death by interfering with the MPP(+)-induced downregulation of mTOR signaling.

5.
Cell Mol Neurobiol ; 33(1): 147-54, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23053546

ABSTRACT

Over the past decade, intense focus has been dedicated on investigating processes involved in the proteolysis of amyloid precursor protein (AßPP) and ß-amyloid (Aß) peptide metabolism, as possible targets for Alzheimer's disease (AD) therapy. To this goal, considerable research has been targeted on potential therapeutic use of compounds promoting non-amyloidogenic processing of AßPP. One of these compounds, oleuropein, a polyphenol constituent of extra virgin olive oil exhibiting a wide range of pharmacological properties, was shown to interact non-covalently with Aß, an interaction that might be related to a potential protective role of oleuropein against Aß aggregation. In the present study, it was demonstrated that oleuropein treatment of HEK293 cells stably transfected with the isoform 695 of human AßPP (APP695) leads to markedly elevated levels of sAPPα and to significant reduction of Aß oligomers. These effects were associated with increased activity of matrix metalloproteinase 9 (MMP-9), whereas no significant alterations in the expression of secretases TACE, ADAM-10 or BACE-1 were observed. Similar results were obtained using the human neuroblastoma cell line SK-N-SH. The experimental data reveal an anti-amyloidogenic effect of oleuropein and suggest a possible protective role for oleuropein against AD, extending the spectrum of beneficial properties of this naturally occurring polyphenol.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Antioxidants/pharmacology , Olea , Polyphenols/pharmacology , Pyrans/pharmacology , Amyloid Precursor Protein Secretases/pharmacology , Cell Line, Tumor , Humans , Iridoid Glucosides , Iridoids
6.
J Neurochem ; 121(2): 239-51, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22192143

ABSTRACT

Evidence accumulating during the past few years points to a significant role of matrix metalloproteinase 9 (MMP9) enzymatic activity in synaptic plasticity and cognitive processes. We have previously demonstrated that MMP9 is involved in receptor-mediated α-secretase-like cleavage of APP in vitro, resulting in increased secretion of sAPPα, the soluble N-terminal product of the non-amyloidogenic pathway known to be involved in neuronal plasticity and memory formation. To study the in vivo role of MMP9, we have generated transgenic mice over-expressing MMP9 in the brain. Herein, we demonstrate that MMP9 transgenic animals display enhanced performance in the non-spatial novel object recognition and the spatial water-maze task and that their enhanced performance was accompanied by increased dendritic spine density in the hippocampus and cortex following behavioural testing. Consistent with the above observations, the electrophysiological analysis revealed prolonged maintenance of long-term synaptic potentiation in hippocampal slices from MMP9 transgenic mice. Moreover, elevated sAPPα levels in the hippocampus and cortex of MPP9 transgenic animals were also observed. Overall, our results extend previous findings on the physiological role of MMP9 in neuronal plasticity and furthermore reveal that, APP may be one of the physiological proteolytic targets of MMP9 in vivo.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Peptide Fragments/metabolism , Animals , Blotting, Western , Brain/enzymology , Brain/physiology , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cognition/physiology , DNA/genetics , Dendritic Spines/physiology , Electrophysiological Phenomena , Exploratory Behavior/physiology , Female , Fluorescent Antibody Technique , Hippocampus/cytology , Hippocampus/physiology , Humans , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Psychomotor Performance/physiology , Real-Time Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor beta/genetics , Recognition, Psychology/physiology
7.
J Alzheimers Dis ; 24(4): 705-19, 2011.
Article in English | MEDLINE | ID: mdl-21321391

ABSTRACT

Amyloid-ß protein precursor (AßPP) is a ubiquitously expressed glycoprotein, which under physiological conditions can be cleaved following two alternative routes; the non-amyloidogenic and the amyloidogenic pathway. Shift of AßPP processing in favor of the amyloidogenic pathway is a key event in the pathogenesis of Alzheimer's disease (AD). Among the factors that regulate AßPP processing, nerve growth factor (NGF) appears to play an important role; abnormal NGF signaling has been implicated in the onset of AD. In the present study, we used PC12 cells to study the effects of NGF on AßPP processing and provide evidence that NGF, through binding to its high affinity receptor, TrkA moderately down-regulates the expression of the ß-secretase ß-site AßPP cleaving enzyme-1 and, most importantly, upregulates the expression of two enzymes with α-secretase activity, a disintegrin and metalloprotease-17 and to a greater extent matrix metalloproteinase-9 (MMP9) in a phosphoinositide kinase-3 dependent manner. Finally, we demonstrate that MMP9 actively participates in NGF-induced α-secretase cleavage of AßPP, thus it contributes to the shift of AßPP processing towards the non-amyloidogenic pathway precluding the formation of neurotoxic Aß peptides.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Matrix Metalloproteinase 9/metabolism , Nerve Growth Factor/physiology , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/toxicity , Amyloid beta-Protein Precursor/biosynthesis , Animals , Down-Regulation/physiology , Nerve Growth Factor/metabolism , PC12 Cells , Protein Binding/drug effects , Protein Binding/physiology , Rats , Signal Transduction/physiology , Up-Regulation/physiology
8.
Proc Natl Acad Sci U S A ; 107(48): 20816-21, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21076037

ABSTRACT

Scavenger receptor class B type I (SR-BI) is a high-density lipoprotein receptor that regulates cholesterol efflux from the peripheral tissues to the liver. SR-BI has been identified on astrocytes and vascular smooth muscle cells in Alzheimer's disease brain and has been shown to mediate adhesion of microglia to fibrillar amyloid-ß (Aß). Here we report that SR-BI mediates perivascular macrophage response and regulates Aß-related pathology and cerebral amyloid angiopathy in an Alzheimer's mouse model. Reduction or deletion of SR-BI gene in heterozygous or homozygous deficient mice (SR-BI(+/-), (-/-)) resulted in a significant increase in perivascular macrophages in the brain. SR-BI deletion had no effect on apolipoprotein E or apolipoprotein AI levels in the mouse brain. Our analysis revealed increased levels of SR-BI expression in the brains of human amyloid precursor protein (Swedish, Indiana) transgenic mice (J20 line). To evaluate the role of SR-BI in Alzheimer's disease pathogenesis, we inactivated one SR-BI allele in J20 transgenic mice. SR-BI reduction in J20/SR-BI(+/-) mice enhanced fibrillar amyloid deposition and cerebral amyloid angiopathy and also exacerbated learning and memory deficits compared with J20 littermates. Immunohistochemical analysis revealed localization of SR-BI on perivascular macrophages in tight association with Aß deposits. Our data suggest that SR-BI reduction impairs the response of perivascular macrophages to Aß and enhances the Aß-related phenotype and cerebral amyloid angiopathy in J20 mice. These results reveal that SR-BI, a scavenger receptor primarily involved in high-density lipoprotein cholesterol transport, plays an essential role in Alzheimer's disease and cerebral amyloid angiopathy.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Blood Vessels/metabolism , Brain/blood supply , Macrophages/metabolism , Scavenger Receptors, Class B/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Apolipoprotein A-I/metabolism , Apolipoproteins E/metabolism , Astrocytes/metabolism , Behavior, Animal , Blood Vessels/pathology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Mice , Microglia/metabolism , Oxidative Stress , Phagocytosis , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Processing, Post-Translational , Protein Transport
9.
Development ; 136(22): 3841-51, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19855026

ABSTRACT

Coordination of voluntary motor activity depends on the generation of the appropriate neuronal subtypes in the basal ganglia and their integration into functional neuronal circuits. The largest nucleus of the basal ganglia, the striatum, contains two classes of neurons: the principal population of medium-sized dense spiny neurons (MSNs; 97-98% of all striatal neurons in rodents), which project to the globus pallidus and the substantia nigra, and the locally projecting striatal interneurons (SINs; 2-3% in rodents). SINs are further subdivided into two non-overlapping groups: those producing acetylcholine (cholinergic) and those producing gamma-amino butyric acid (GABAergic). Despite the pivotal role of SINs in integrating the output of striatal circuits and the function of neuronal networks in the ventral forebrain, the lineage relationship of SIN subtypes and the molecular mechanisms that control their differentiation are currently unclear. Using genetic fate mapping, we demonstrate here that the majority of cholinergic and GABAergic SINs are derived from common precursors generated in the medial ganglionic eminence during embryogenesis. These precursors express the LIM homeodomain protein Lhx6 and have characteristics of proto-GABAergic neurons. By combining gene expression analysis with loss-of-function and misexpression experiments, we provide evidence that the differentiation of the common precursor into mature SIN subtypes is regulated by the combinatorial activity of the LIM homeodomain proteins Lhx6, Lhx7 (Lhx8) and Isl1. These studies suggest that a LIM homeodomain transcriptional code confers cell-fate specification and neurotransmitter identity in neuronal subpopulations of the ventral forebrain.


Subject(s)
Homeodomain Proteins/metabolism , Interneurons/cytology , Prosencephalon/embryology , Animals , Embryo, Mammalian , Ganglia/metabolism , Mice , Prosencephalon/cytology
10.
Eur J Neurosci ; 21(11): 2923-38, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15978004

ABSTRACT

The identification of the genetic determinants specifying neuronal networks in the mammalian brain is crucial for the understanding of the molecular and cellular mechanisms that ultimately control cognitive functions. Here we have generated a targeted allele of the LIM-homeodomain-encoding gene Lhx7 by replacing exons 3-5 with a LacZ reporter. In heterozygous animals, which are healthy, fertile and have no apparent cellular deficit in the forebrain, b-galactosidase activity reproduces the pattern of expression of the wild-type Lhx7 locus. However, homozygous mutant mice show severe deficits in forebrain cholinergic neurons (FCNs), while other classes of forebrain neurons appear unaffected. Using the LacZ reporter as a marker, we show that in LHX7-deficient mice FCN progenitors survive but fail to generate cholinergic interneurons in the striatum and cholinergic projection neurons in the basal forebrain. Analysis of behaviour in a series of spatial and non-spatial learning and memory tasks revealed that FCN ablation in Lhx7 mutants is associated with severe deficits in spatial but only mild impairment of non-spatial learning and memory. In addition, we found no deficit in long-term potentiation in mutant animals, suggesting that FCNs modulate hippocampal function independently of its capacity to store information. Overall our experiments demonstrate that Lhx7 expression is required for the specification or differentiation of cholinergic forebrain neurons involved in the processing of spatial information.


Subject(s)
Basal Nucleus of Meynert/abnormalities , Basal Nucleus of Meynert/metabolism , Cholinergic Fibers/metabolism , Homeodomain Proteins/genetics , Learning Disabilities/metabolism , Memory Disorders/metabolism , Prosencephalon/metabolism , Acetylcholine/metabolism , Animals , Basal Nucleus of Meynert/pathology , Cell Death/genetics , Cell Differentiation/genetics , Cholinergic Fibers/pathology , Corpus Striatum/abnormalities , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dentate Gyrus/metabolism , Dentate Gyrus/physiopathology , Disease Models, Animal , Genes, Reporter/genetics , Homeodomain Proteins/metabolism , Interneurons/metabolism , Interneurons/pathology , LIM-Homeodomain Proteins , Lac Operon/genetics , Learning Disabilities/genetics , Learning Disabilities/physiopathology , Long-Term Potentiation/genetics , Male , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice , Mice, Knockout , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nervous System Malformations/physiopathology , Prosencephalon/physiopathology , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...