Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167187, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653354

ABSTRACT

Clinical relevance of miRNAs as biomarkers is growing due to their stability and detection in biofluids. In this, diagnosis at asymptomatic stages of Alzheimer's disease (AD) remains a challenge since it can only be made at autopsy according to Braak NFT staging. Achieving the objective of detecting AD at early stages would allow possible therapies to be addressed before the onset of cognitive impairment. Many studies have determined that the expression pattern of some miRNAs is dysregulated in AD patients, but to date, none has been correlated with downregulated expression of cellular prion protein (PrPC) during disease progression. That is why, by means of cross studies of miRNAs up-regulated in AD with in silico identification of potential miRNAs-binding to 3'UTR of human PRNP gene, we selected miR-519a-3p for our study. Then, in vitro experiments were carried out in two ways. First, we validated miR-519a-3p target on 3'UTR-PRNP, and second, we analyzed the levels of PrPC expression after using of mimic technology on cell culture. In addition, RT-qPCR was performed to analyzed miR-519a-3p expression in human cerebral samples of AD at different stages of disease evolution. Additionally, samples of other neurodegenerative diseases such as other non-AD tauopathies and several synucleinopathies were included in the study. Our results showed that miR-519a-3p overlaps with PRNP 3'UTR in vitro and promotes downregulation of PrPC. Moreover, miR-519a-3p was found to be up-regulated exclusively in AD samples from stage I to VI, suggesting its potential use as a novel label of preclinical stages of the disease.


Subject(s)
3' Untranslated Regions , Alzheimer Disease , Biomarkers , MicroRNAs , Prion Proteins , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/diagnosis , Prion Proteins/genetics , Prion Proteins/metabolism , Biomarkers/metabolism , 3' Untranslated Regions/genetics , Female , Aged , Male , Aged, 80 and over , PrPC Proteins/metabolism , PrPC Proteins/genetics
2.
Front Mol Neurosci ; 16: 1253954, 2023.
Article in English | MEDLINE | ID: mdl-37829513

ABSTRACT

Introduction: Brain connectivity requires correct axonal guidance to drive axons to their appropriate targets. This process is orchestrated by guidance cues that exert attraction or repulsion to developing axons. However, the intricacies of the cellular machinery responsible for the correct response of growth cones are just being unveiled. Netrin-1 is a bifunctional molecule involved in axon pathfinding and cell migration that induces repulsion during postnatal cerebellar development. This process is mediated by UNC5 homolog receptors located on external granule layer (EGL) tracts. Methods: Biochemical, imaging and cell biology techniques, as well as syntaxin-1A/B (Stx1A/B) knock-out mice were used in primary cultures and brain explants. Results and discussion: Here, we demonstrate that this response is characterized by enhanced membrane internalization through macropinocytosis, but not clathrin-mediated endocytosis. We show that UNC5A, UNC5B, and UNC5C receptors form a protein complex with the t-SNARE syntaxin-1. By combining botulinum neurotoxins, an shRNA knock-down strategy and Stx1 knock-out mice, we demonstrate that this SNARE protein is required for Netrin1-induced macropinocytosis and chemorepulsion, suggesting that Stx1 is crucial in regulating Netrin-1-mediated axonal guidance.

3.
Front Neuroanat ; 13: 3, 2019.
Article in English | MEDLINE | ID: mdl-30833889

ABSTRACT

The metallic impregnation invented by Camillo Golgi in 1873 has allowed the visualization of individual neurons in their entirety, leading to a breakthrough in the knowledge on the structure of the nervous system. Professor of Histology and of General Pathology, Golgi worked for decades at the University of Pavia, leading a very active laboratory. Unfortunately, most of Golgi's histological preparations are lost. The present contribution provides an account of the original slides on the nervous system from Golgi's laboratory available nowadays at the Golgi Museum and Historical Museum of the University of Pavia. Knowledge on the organization of the nervous tissue at the time of Golgi's observations is recalled. Notes on the equipment of Golgi's laboratory and the methodology Golgi used for his preparations are presented. Images of neurons from his slides (mostly from hippocampus, neocortex and cerebellum) are here shown for the first time together with some of Golgi's drawings. The sections are stained with the Golgi impregnation and Cajal stain. Golgi-impregnated sections are very thick (some more than 150 µm) and require continuous focusing during the microscopic observation. Heterogeneity of neuronal size and shape, free endings of distal dendritic arborizations, axonal branching stand out at the microscopic observation of Golgi-impregnated sections and in Golgi's drawings, and were novel findings at his time. Golgi also pointed out that the axon only originates from cell bodies, representing a constant and distinctive feature of nerve cells which distinguishes them from glia, and subserving transmission at a distance. Dendritic spines can be seen in some cortical neurons, although Golgi, possibly worried about artifacts, did not identify them. The puzzling intricacy of fully impregnated nervous tissue components offered to the first microscopic observations still elicit nowadays the emotion Golgi must have felt looking at his slides.

4.
Oncotarget ; 9(89): 35922-35940, 2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30542508

ABSTRACT

SNARE proteins are essential components of the machinery that regulates vesicle trafficking and exocytosis. Their role is critical for the membrane-fusion processes that occur during neurotransmitter release. However, research in the last decade has also unraveled the relevance of these proteins in membrane expansion and cytoskeletal rearrangements during developmental processes such as neuronal migration and growth cone extension and attraction. Neurotrophins are neurotrophic factors that are required for many cellular functions throughout the brain, including neurite outgrowth and guidance, synaptic formation, and plasticity. Here we show that neurotrophin Trk receptors form a specific protein complex with the t-SNARE protein Syntaxin 1, both in vivo and in vitro. We also demonstrate that blockade of Syntaxin 1 abolishes neurotrophin-dependent growth of axons in neuronal cultures and decreases exocytotic events at the tip of axonal growth cones. 25-kDa soluble N-ethylmaleimide-sensitive factor attachment protein and Vesicle-associated membrane protein 2 do not participate in the formation of this SNARE complex, while tetanus neurotoxin-insensitive vesicle-associated membrane protein interacts with Trk receptors; knockdown of this (v) SNARE impairs Trk-dependent outgrowth. Taken together, our results support the notion that an atypical SNARE complex comprising Syntaxin 1 and tetanus neurotoxin-insensitive vesicle-associated membrane protein is required for axonal neurotrophin function.

5.
PLoS Genet ; 14(6): e1007432, 2018 06.
Article in English | MEDLINE | ID: mdl-29912942

ABSTRACT

Axonal growth and guidance rely on correct growth cone responses to guidance cues. Unlike the signaling cascades that link axonal growth to cytoskeletal dynamics, little is known about the crosstalk mechanisms between guidance and membrane dynamics and turnover. Recent studies indicate that whereas axonal attraction requires exocytosis, chemorepulsion relies on endocytosis. Indeed, our own studies have shown that Netrin-1/Deleted in Colorectal Cancer (DCC) signaling triggers exocytosis through the SNARE Syntaxin-1 (STX1). However, limited in vivo evidence is available about the role of SNARE proteins in axonal guidance. To address this issue, here we systematically deleted SNARE genes in three species. We show that loss-of-function of STX1 results in pre- and post-commissural axonal guidance defects in the midline of fly, chick, and mouse embryos. Inactivation of VAMP2, Ti-VAMP, and SNAP25 led to additional abnormalities in axonal guidance. We also confirmed that STX1 loss-of-function results in reduced sensitivity of commissural axons to Slit-2 and Netrin-1. Finally, genetic interaction studies in Drosophila show that STX1 interacts with both the Netrin-1/DCC and Robo/Slit pathways. Our data provide evidence of an evolutionarily conserved role of STX1 and SNARE proteins in midline axonal guidance in vivo, by regulating both pre- and post-commissural guidance mechanisms.


Subject(s)
Neurogenesis/genetics , Syntaxin 1/genetics , Syntaxin 1/physiology , Animals , Axons/metabolism , Chemotaxis/genetics , Chick Embryo , Drosophila/genetics , Drosophila Proteins/genetics , Exocytosis/genetics , Gene Expression Regulation, Developmental/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Netrin-1/genetics , Netrin-1/metabolism , Neurogenesis/physiology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/physiology , SNARE Proteins/genetics , SNARE Proteins/metabolism , Signal Transduction/genetics , Spinal Cord/embryology , Spinal Cord/metabolism
6.
Neural Regen Res ; 13(3): 386-392, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29623913

ABSTRACT

Through complex mechanisms that guide axons to the appropriate routes towards their targets, axonal growth and guidance lead to neuronal system formation. These mechanisms establish the synaptic circuitry necessary for the optimal performance of the nervous system in all organisms. Damage to these networks can be repaired by neuroregenerative processes which in turn can re-establish synapses between injured axons and postsynaptic terminals. Both axonal growth and guidance and the neuroregenerative response rely on correct axonal growth and growth cone responses to guidance cues as well as correct synapses with appropriate targets. With this in mind, parallels can be drawn between axonal regeneration and processes occurring during embryonic nervous system development. However, when studying parallels between axonal development and regeneration many questions still arise; mainly, how do axons grow and synapse with their targets and how do they repair their membranes, grow and orchestrate regenerative responses after injury. Major players in the cellular and molecular processes that lead to growth cone development and movement during embryonic development are the Soluble N-ethylamaleimide Sensitive Factor (NSF) Attachment Protein Receptor (SNARE) proteins, which have been shown to be involved in axonal growth and guidance. Their involvement in axonal growth, guidance and neuroregeneration is of foremost importance, due to their roles in vesicle and membrane trafficking events. Here, we review the recent literature on the involvement of SNARE proteins in axonal growth and guidance during embryonic development and neuroregeneration.

7.
Front Neuroanat ; 12: 13, 2018.
Article in English | MEDLINE | ID: mdl-29535612

ABSTRACT

Neuron populations of the lateral hypothalamus which synthesize the orexin (OX)/hypocretin or melanin-concentrating hormone (MCH) peptides play crucial, reciprocal roles in regulating wake stability and sleep. The disease human African trypanosomiasis (HAT), also called sleeping sickness, caused by extracellular Trypanosoma brucei (T. b.) parasites, leads to characteristic sleep-wake cycle disruption and narcoleptic-like alterations of the sleep structure. Previous studies have revealed damage of OX and MCH neurons during systemic infection of laboratory rodents with the non-human pathogenic T. b. brucei subspecies. No information is available, however, on these peptidergic neurons after systemic infection with T. b. gambiense, the etiological agent of 97% of HAT cases. The present study was aimed at the investigation of immunohistochemically characterized OX and MCH neurons after T. b. gambiense or T. b. brucei infection of a susceptible rodent, the multimammate mouse, Mastomysnatalensis. Cell counts and evaluation of OX fiber density were performed at 4 and 8 weeks post-infection, when parasites had entered the brain parenchyma from the periphery. A significant decrease of OX neurons (about 44% reduction) and MCH neurons (about 54% reduction) was found in the lateral hypothalamus and perifornical area at 8 weeks in T. b. gambiense-infected M. natalensis. A moderate decrease (21% and 24% reduction, respectively), which did not reach statistical significance, was found after T. b. brucei infection. In two key targets of diencephalic orexinergic innervation, the peri-suprachiasmatic nucleus (SCN) region and the thalamic paraventricular nucleus (PVT), densitometric analyses showed a significant progressive decrease in the density of orexinergic fibers in both infection paradigms, and especially during T. b. gambiense infection. Altogether the findings provide novel information showing that OX and MCH neurons are highly vulnerable to chronic neuroinflammatory signaling caused by the infection of human-pathogenic African trypanosomes.

8.
Ann Transplant ; 23: 144-152, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29483486

ABSTRACT

BACKGROUND Heart transplantation is a therapeutic option for patients with severe coronary artery disease or heart failure. One of the difficulties to overcome is the apoptosis of cardiomyocytes in the donor organ. To prevent apoptosis in the donor organ, we developed a fusion protein containing FLIP (FADD-like interleukin beta-converting enzyme (FLICE)-like inhibitory protein) to inhibit caspase-8. MATERIAL AND METHODS We linked the cDNA coding for the FLIP protein to the transduction domain of HIV (human immunodeficiency virus) to allow the protein to enter cells. The recombinant protein was used at two different concentrations, 3 nM and 30 nM, for treatment of the donor heart in rat transplantation experiments. After transplantation, apoptosis was measured by ELISA, and the levels of active caspase-3, caspase-8, Bid, and PUMA were determined by western blotting using specific antibodies. RESULTS We observed that treatment of the donor organ with a solution containing this protein reduced the apoptosis level in the donor organ after 30 minutes post-transplantation as measured by the total of apoptotic cells with ELISA assay, and caspase-8 and caspase-3 activation and decreased levels of BH3-only proteins such as Bid and PUMA. Furthermore, this treatment also reduced the total tyrosine phosphorylation levels, which may be a possible measurement of lower oxidative stress levels in cardiomyocytes. CONCLUSIONS Protein FLIP solution reduced apoptosis at 30 minutes post-transplantation and decreased levels of several regulators of apoptosis.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/administration & dosage , Cardioplegic Solutions , Heart Transplantation/methods , Heart/drug effects , Animals , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Male , Myocardium/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
10.
Dev Neurobiol ; 77(8): 963-974, 2017 09.
Article in English | MEDLINE | ID: mdl-28033683

ABSTRACT

Axonal growth and guidance rely on correct growth cone responses to guidance cues, both in the central nervous system (CNS) and in the periphery. Unlike the signaling cascades that link axonal growth to cytoskeletal dynamics, little is known about the cross-talk mechanisms between guidance and membrane dynamics and turnover in the axon. Our studies have shown that Netrin-1/deleted in colorectal cancer signaling triggers exocytosis through the SNARE Syntaxin-1 (STX-1) during the formation of commissural pathways. However, limited in vivo evidence is available about the role of SNARE proteins in motor axonal guidance. Here we show that loss-of-function of SNARE complex members results in motor axon guidance defects in fly and chick embryos. Knock-down of Syntaxin-1, VAMP-2, and SNAP-25 leads to abnormalities in the motor axon routes out of the CNS. Our data point to an evolutionarily conserved role of the SNARE complex proteins in motor axon guidance, thereby pinpointing an important function of SNARE proteins in axonal navigation in vivo. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 963-974, 2017.


Subject(s)
Avian Proteins/metabolism , Axons/metabolism , Drosophila Proteins/metabolism , Motor Neurons/metabolism , Neuronal Outgrowth/physiology , SNARE Proteins/metabolism , Animals , Chick Embryo , Drosophila melanogaster , Immunohistochemistry , Species Specificity
11.
J Neurosci ; 35(13): 5156-70, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25834042

ABSTRACT

Axonal guidance and synaptic specification depends on specific signaling mechanisms that occur in growth cones. While several signaling pathways implicated in cone navigation have been identified, membrane dynamics in growth cones remains largely unknown. We took advantage of SynaptopHluorin and high-speed optical recordings to monitor the patterns of membrane dynamics in rat hippocampal growth cones. We show that exocytosis occurs both at the peripheral and central domains, including filopodia, and that SynaptopHluorin signals occur as spontaneous patterned peaks. Such transients average approximately two per minute and last ∼30 s. We also demonstrate that the chemoattractant Netrin-1 elicits increases in the frequency and slopes of these transients, with peaks averaging up to six per minute in the peripheral domain. Netrin-1-dependent regulation of exocytotic events requires the activation of the Erk1/2 and SFK pathways. Furthermore, we show that domains with high SynaptopHluorin signals correlate with high local calcium concentrations and that local, spontaneous calcium increases are associated with higher SynaptopHluorin signals. These findings demonstrate highly stereotyped, spontaneous transients of local exocytosis in growth cones and that these transients are positively regulated by chemoattractant molecules such as Netrin-1.


Subject(s)
Exocytosis/physiology , Growth Cones/physiology , Nerve Growth Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Calcium Signaling/physiology , Cells, Cultured , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Hippocampus/physiology , Netrin-1 , Neurons/physiology , Optical Imaging , Rats , Recombinant Fusion Proteins/metabolism
12.
PLoS One ; 10(3): e0119707, 2015.
Article in English | MEDLINE | ID: mdl-25803850

ABSTRACT

Glioblastoma (GBM) is the most prevalent adult brain tumor, with virtually no cure, and with a median overall survival of 15 months from diagnosis despite of the treatment. SNARE proteins mediate membrane fusion events in cells and are essential for many cellular processes including exocytosis and neurotransmission, intracellular trafficking and cell migration. Here we show that the blockade of the SNARE protein Syntaxin 1 (Stx1) function impairs GBM cell proliferation. We show that Stx1 loss-of-function in GBM cells, through ShRNA lentiviral transduction, a Stx1 dominant negative and botulinum toxins, dramatically reduces the growth of GBM after grafting U373 cells into the brain of immune compromised mice. Interestingly, Stx1 role on GBM progression may not be restricted just to cell proliferation since the blockade of Stx1 also reduces in vitro GBM cell invasiveness suggesting a role in several processes relevant for tumor progression. Altogether, our findings indicate that the blockade of SNARE proteins may represent a novel therapeutic tool against GBM.


Subject(s)
Botulinum Toxins/pharmacology , Cell Proliferation/drug effects , Glioblastoma/physiopathology , RNA, Small Interfering/pharmacology , Syntaxin 1/antagonists & inhibitors , Animals , Blotting, Western , Bromodeoxyuridine , Cell Line, Tumor , Flow Cytometry , Glioblastoma/drug therapy , Humans , Lentivirus , Mice , Neoplasm Invasiveness/prevention & control , RNA, Small Interfering/genetics , Statistics, Nonparametric , Transduction, Genetic/methods
13.
Eur J Neurosci ; 36(9): 3152-64, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22946563

ABSTRACT

Directed cell migration and axonal guidance are essential steps in neural development that share many molecular mechanisms. The guidance of developing axons and migrating neurons is likely to depend on the precise control of plasmalemma turnover in selected regions of leading edges and growth cones, respectively. Previous results provided evidence of a signaling mechanism that couples chemotropic deleted in colorectal cancer (DCC)/Netrin-1 axonal guidance and exocytosis through Syntaxin1(Sytx1)/TI-VAMP SNARE proteins. Here we studied whether Netrin-1-dependent neuronal migration relies on a similar SNARE mechanism. We show that migrating neurons in the lower rhombic lip (LRL) express several SNARE proteins, and that DCC co-associates with Sytx1 and TI-VAMP in these cells. We also demonstrate that cleavage of Sytx1 by botulinum toxin C1 (BoNT/C1) abolishes Netrin-1-dependent chemoattraction of migrating neurons, and that interference of Sytx1 functions with shRNAs or Sytx1-dominant negatives disrupts Netrin-1-dependent chemoattraction of LRL neurons. These findings indicate that a Sytx1/DCC interaction is required for Netrin-1 guidance of migrating neurons, thereby highlighting a relationship between guidance signaling and SNARE proteins that regulate membrane turnover.


Subject(s)
Cerebellum/metabolism , Chemotaxis , Nerve Growth Factors/metabolism , Neurons/metabolism , Receptors, Cell Surface/metabolism , Syntaxin 1/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Botulinum Toxins/pharmacology , Cerebellum/cytology , Cerebellum/embryology , Chemotaxis/drug effects , Chemotaxis/genetics , DCC Receptor , Gene Expression Regulation, Developmental , Mice , Nerve Growth Factors/antagonists & inhibitors , Netrin-1 , RNA, Small Interfering , Receptors, Cell Surface/genetics , Signal Transduction , Syntaxin 1/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Vesicle-Associated Membrane Protein 2/metabolism
14.
J Neurosci ; 31(41): 14463-80, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21994363

ABSTRACT

Directed cell migration and axonal guidance are essential steps in neural development. Both processes are controlled by specific guidance cues that activate the signaling cascades that ultimately control cytoskeletal dynamics. Another essential step in migration and axonal guidance is the regulation of plasmalemma turnover and exocytosis in leading edges and growth cones. However, the cross talk mechanisms linking guidance receptors and membrane exocytosis are not understood. Netrin-1 is a chemoattractive cue required for the formation of commissural pathways. Here, we show that the Netrin-1 receptor deleted in colorectal cancer (DCC) forms a protein complex with the t-SNARE (target SNARE) protein Syntaxin-1 (Sytx1). This interaction is Netrin-1 dependent both in vitro and in vivo, and requires specific Sytx1 and DCC domains. Blockade of Sytx1 function by using botulinum toxins abolished Netrin-1-dependent chemoattraction of axons in mouse neuronal cultures. Similar loss-of-function experiments in the chicken spinal cord in vivo using dominant-negative Sytx1 constructs or RNAi led to defects in commissural axon pathfinding reminiscent to those described in Netrin-1 and DCC loss-of-function models. We also show that Netrin-1 elicits exocytosis at growth cones in a Sytx1-dependent manner. Moreover, we demonstrate that the Sytx1/DCC complex associates with the v-SNARE (vesicle SNARE) tetanus neurotoxin-insensitive vesicle-associated membrane protein (TI-VAMP) and that knockdown of TI-VAMP in the commissural pathway in the spinal cord results in aberrant axonal guidance phenotypes. Our data provide evidence of a new signaling mechanism that couples chemotropic Netrin-1/DCC axonal guidance and Sytx1/TI-VAMP SNARE proteins regulating membrane turnover and exocytosis.


Subject(s)
Exocytosis/physiology , Growth Cones/physiology , Nerve Growth Factors/metabolism , Neurons/cytology , Receptors, Cell Surface/metabolism , SNARE Proteins/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins/metabolism , Analysis of Variance , Animals , Animals, Newborn , Axons/drug effects , Axons/physiology , Boron Compounds/metabolism , Botulinum Toxins, Type A/pharmacology , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Chemotaxis/drug effects , Chlorocebus aethiops , Complement C1/pharmacology , DCC Receptor , Embryo, Mammalian , Exocytosis/drug effects , Exocytosis/genetics , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Growth Cones/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/cytology , Humans , Immunoprecipitation , Mice , Mice, Knockout , Mice, Transgenic , Munc18 Proteins/genetics , Munc18 Proteins/metabolism , Nerve Growth Factors/genetics , Nerve Tissue Proteins/metabolism , Netrin-1 , Neuromuscular Agents/pharmacology , Organ Culture Techniques , Receptors, Cell Surface/genetics , SNARE Proteins/genetics , Signal Transduction/drug effects , Surface Plasmon Resonance/methods , Tetanus Toxin/pharmacology , Transfection/methods , Tumor Suppressor Proteins/genetics , Vesicle-Associated Membrane Protein 2/metabolism
15.
Neurobiol Dis ; 37(3): 682-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20025970

ABSTRACT

Reelin is a glycoprotein that modulates synaptic function and plasticity in the mature brain, thereby favouring memory formation. We recently reported altered cerebral Reelin expression in Alzheimer's disease (AD). Here we demonstrate pronounced Reelin changes at protein and mRNA levels in the frontal cortex in adult Down's syndrome (DS), where the extra copy of chromosome 21 leads to overexpression of beta-amyloid. In cortical extracts of fetal DS samples we detected increased levels of the full-length Reelin and the 310-kDa fragment. Overexpression of mutant human amyloid precursor protein also led to an increase in levels of Reelin fragments in Tg2576 transgenic mice for human beta-amyloid. Finally, in vitro Abeta42 treatment of SH-SY5Y neuroblastoma cells led to increased Reelin levels. An altered pattern of Reelin glycosylation was detected in extracts from the frontal cortex of AD patients and in Abeta42-treated SH-SY5Y cells, supporting the notion that beta-amyloid triggers altered Reelin processing. These results provide evidence that Reelin expression and processing is altered in several amyloid conditions.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Adult , Aged , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/physiopathology , Cell Adhesion Molecules, Neuronal/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Down Syndrome/metabolism , Down Syndrome/physiopathology , Extracellular Matrix Proteins/genetics , Female , Fetus , Gene Expression Regulation/physiology , Glycosylation , Humans , Male , Mice , Middle Aged , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , RNA, Messenger/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Synapses/metabolism , Up-Regulation/genetics
16.
J Neurosci ; 23(9): 3566-71, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12736326

ABSTRACT

Total lack of visual experience [dark rearing (DR)] is known to prolong the critical period and delay development of sensory functions in mammalian visual cortex. Recent results show that neurotrophins (NTs) counteract the effects of DR on functional properties of visual cortical cells and exert a strong control on critical period duration. NTs are known to modulate the development and synaptic efficacy of neurotransmitter systems that are affected by DR. However, it is still unknown whether the actions of NTs in dark-reared animals involve interaction with neurotransmitter systems. We have studied the effects of DR on the expression of key molecules in the glutamatergic and GABAergic systems in control and NT-treated animals. We have found that DR reduced the expression of the NMDA receptor 2A subunit and its associated protein PSD-95 (postsynaptic density-95), of GRIP (AMPA glutamate receptor interacting protein), and of the biosynthetic enzyme GAD (glutamic acid decarboxylase). Returning dark-reared animals to light for 2 hr restored normal expression of the above-mentioned proteins almost completely. NT treatment specifically counteracts DR effects; NGF acts primarily on the NMDA system, whereas BDNF acts primarily on the GABAergic system. Finally, the action of NT4 seems to involve both excitatory and inhibitory systems. These data demonstrate that different NTs counteract DR effects by modulating the expression of key molecules of the excitatory and inhibitory neurotransmitter systems.


Subject(s)
Darkness , Nerve Growth Factors/pharmacology , Proteins/metabolism , Synapses/metabolism , Visual Cortex/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Carrier Proteins/metabolism , Densitometry , Disks Large Homolog 4 Protein , Glutamate Decarboxylase/metabolism , Immunoblotting , Intracellular Signaling Peptides and Proteins , Isoenzymes/metabolism , Light , Membrane Proteins , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , Photic Stimulation , Rats , Rats, Long-Evans , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Sensory Deprivation/physiology , Synapses/drug effects , Visual Cortex/drug effects
17.
Eur J Neurosci ; 16(10): 1925-30, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12453056

ABSTRACT

Total lack of visual experience (dark rearing, DR) is known to affect development of mammalian visual cortex (VC) and to prolong the critical period of visual cortical plasticity. Neurotrophins (NTs) have been proposed to play a relevant role in activity dependent processes important for the final shaping of cortical visual connections. Neurotrophin supply or antagonism of endogenous NT action profoundly affect visual cortical development and plasticity; in particular, exogenous supply of NTs counteracts DR effects on VC development. However, the effects of DR on NT expression are still debated and mounting evidence reports a mismatch between BDNF mRNA and protein expression in DR animals. To gain insight into the effects of DR on expression of nerve growth factor (NGF) and the functional state of NT signalling pathways, we assessed the phosphorylation state of Trk receptors in light-reared animals (LR), in dark-reared animals (DR), in DR animals briefly exposed to light and in DR animals with exogenous supply of NTs [NGF, brain-derived neurotrophic factor (BDNF) and NT-4] in the VC. We report that DR increases the expression of NGF but reduces the phosphorylation of TrkA and TrkB receptors with respect to LR; normal phosphorylation is rapidly rescued by a brief exposure to light. Exogenous supply of NGF, BDNF or NT4 in DR animals also rescues the phosphorylation of their receptors.


Subject(s)
Darkness/adverse effects , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Sensory Deprivation , Visual Cortex/metabolism , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/pharmacology , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Nerve Growth Factor/pharmacology , Nerve Growth Factors/pharmacology , Phosphorylation/drug effects , Photic Stimulation , Precipitin Tests , Rats , Rats, Long-Evans , Receptor, trkA/drug effects , Receptor, trkB/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Visual Cortex/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...