Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
J Neurosci ; 37(3): 546-561, 2017 01 18.
Article in English | MEDLINE | ID: mdl-28100738

ABSTRACT

MicroRNAs (miRNA) play an important role in post-transcriptional gene regulation of several physiological and pathological processes. In multiple sclerosis (MS), a chronic inflammatory and degenerative disease of the CNS, and in its mouse model, the experimental autoimmune encephalomyelitis (EAE), miRNA dysregulation has been mainly related to immune system dysfunction and white matter (WM) pathology. However, little is known about their role in gray matter pathology. Here, we explored miRNA involvement in the inflammation-driven alterations of synaptic structure and function, collectively known as synaptopathy, a neuropathological process contributing to excitotoxic neurodegeneration in MS/EAE. Particularly, we observed that miR-142-3p is increased in the CSF of patients with active MS and in EAE brains. We propose miR-142-3p as a molecular mediator of the IL-1ß-dependent downregulation of the glial glutamate-aspartate transporter (GLAST), which causes an enhancement of the glutamatergic transmission in the EAE cerebellum. The synaptic abnormalities mediated by IL-1ß and the clinical and neuropathological manifestations of EAE disappeared in miR-142 knock-out mice. Furthermore, we observed that in vivo miR-142-3p inhibition, either by a preventive and local treatment or by a therapeutic and systemic strategy, abolished IL-1ß- and GLAST-dependent synaptopathy in EAE wild-type mice. Consistently, miR-142-3p was responsible for the glutamatergic synaptic alterations caused by CSF of patients with MS, and CSF levels of miR-142-3p correlated with prospective MS disease progression. Our findings highlight miR-142-3p as key molecular player in IL-1ß-mediated synaptic dysfunction, possibly leading to excitotoxic damage in both EAE and MS diseases. Inhibition of miR-142-3p could be neuroprotective in MS. SIGNIFICANCE STATEMENT: Current studies suggest the role of glutamate excitotoxicity in the development and progression of multiple sclerosis (MS) and of its mouse model experimental autoimmune encephalomyelitis (EAE). The molecular mechanisms linking inflammation and synaptic alterations in MS/EAE are still unknown. Here, we identified miR-142-3p as a determinant molecular actor in inflammation-dependent synaptopathy typical of both MS and EAE. miR-142-3p was upregulated in the CSF of MS patients and in EAE cerebellum. Inhibition of miR-142-3p, locally in EAE brain and in a MS chimeric ex vivo model, recovered glutamatergic synaptic enhancement typical of EAE/MS. We proved that miR-142-3p promoted the IL-1ß-dependent glutamate dysfunction by targeting glutamate-aspartate transporter (GLAST), a crucial glial transporter involved in glutamate homeostasis. Finally, we suggest miR-142-3p as a negative prognostic factor in patients with relapsing-remitting multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Interleukin-1beta/biosynthesis , MicroRNAs/biosynthesis , Multiple Sclerosis, Relapsing-Remitting/metabolism , Synapses/metabolism , Adult , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Knock-In Techniques , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/cerebrospinal fluid , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Synapses/pathology
2.
J Cell Physiol ; 233(10): 7178-7187, 2018 10.
Article in English | MEDLINE | ID: mdl-29741791

ABSTRACT

Nerve growth factor, the prototype of a family of neurotrophins, elicits differentiation and survival of peripheral and central neuronal cells. Although its neural mechanisms have been studied extensively, relatively little is known about the transcriptional regulation governing its effects. We have previously observed that in primary cultures of rat hippocampal neurons treatment with nerve growth factor for 72 hr increases neurite outgrowth and cell survival. To obtain a comprehensive view of the underlying transcriptional program, we performed whole-genome expression analysis by microarray technology. We identified 541 differentially expressed genes and characterized dysregulated pathways related to innate immunity: the complement system and neuro-inflammatory signaling. The exploitation of such genes and pathways may help interfering with the intracellular mechanisms involved in neuronal survival and guide novel therapeutic strategies for neurodegenerative diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain-Derived Neurotrophic Factor/drug effects , Inflammation/drug therapy , Neurons/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Survival/drug effects , Cells, Cultured , Gene Expression Regulation/physiology , Hippocampus/drug effects , Immunosuppression Therapy/methods , Neurites/drug effects , Neuronal Outgrowth/drug effects , Neurons/metabolism , Rats, Wistar
3.
J Cell Sci ; 129(4): 804-16, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26743087

ABSTRACT

Disconnection between membrane signalling and actin networks can have catastrophic effects depending on cell size and polarity. The survival motor neuron (SMN) protein is ubiquitously involved in assembly of spliceosomal small nuclear ribonucleoprotein particles. Other SMN functions could, however, affect cellular activities driving asymmetrical cell surface expansions. Genes able to mitigate SMN deficiency operate within pathways in which SMN can act, such as mRNA translation, actin network and endocytosis. Here, we found that SMN accumulates at membrane protrusions during the dynamic rearrangement of the actin filaments. In addition to localization data, we show that SMN interacts with caveolin-1, which mediates anchoring of translation machinery components. Importantly, SMN deficiency depletes the plasma membrane of ribosomes, and this correlates with the failure of fibroblasts to extend membrane protrusions. These findings strongly support a relationship between SMN and membrane dynamics. We propose that SMN could assembly translational platforms associated with and governed by the plasma membrane. This activity could be crucial in cells that have an exacerbated interdependence of membrane remodelling and local protein synthesis.


Subject(s)
Cell Membrane/metabolism , SMN Complex Proteins/physiology , Actin Cytoskeleton/metabolism , Caveolin 1/metabolism , Cell Membrane/ultrastructure , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Cells, Cultured , Humans , Protein Biosynthesis , Protein Transport , Ribosomes/metabolism
4.
Int J Mol Sci ; 18(6)2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28632177

ABSTRACT

Dysfunction of nerve growth factor (NGF) and its high-affinity Tropomyosin receptor kinase A (TrkA) receptor has been suggested to contribute to the selective degeneration of basal forebrain cholinergic neurons (BFCN) associated with the progressive cognitive decline in Alzheimer's disease (AD). The aim of this review is to describe our progress in elucidating the molecular mechanisms underlying the dynamic interplay between NGF/TrkA signaling and amyloid precursor protein (APP) metabolism within the context of AD neuropathology. This is mainly based on the finding that TrkA receptor binding to APP depends on a minimal stretch of ~20 amino acids located in the juxtamembrane/extracellular domain of APP that carries the α- and ß-secretase cleavage sites. Here, we provide evidence that: (i) NGF could be one of the "routing" proteins responsible for modulating the metabolism of APP from amyloidogenic towards non-amyloidogenic processing via binding to the TrkA receptor; (ii) the loss of NGF/TrkA signaling could be linked to sporadic AD contributing to the classical hallmarks of the neuropathology, such as synaptic loss, ß-amyloid peptide (Aß) deposition and tau abnormalities. These findings will hopefully help to design therapeutic strategies for AD treatment aimed at preserving cholinergic function and anti-amyloidogenic activity of the physiological NGF/TrkA pathway in the septo-hippocampal system.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Signal Transduction , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins , Animals , Cholinergic Neurons , Hippocampus/metabolism , Humans , Neuropathology , Synapses/metabolism , tau Proteins/metabolism
5.
Cell Mol Life Sci ; 72(21): 4173-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25975226

ABSTRACT

The establishment of neuronal polarity and axonal outgrowth are key processes affecting neuronal migration and synapse formation, their impairment likely leading to cognitive deficits. Here we have found that the apoptotic protease activating factor 1 (Apaf1), apart from its canonical role in apoptosis, plays an additional function in cortical neurons, where its deficiency specifically impairs axonal growth. Given the central role played by centrosomes and microtubules in the polarized extension of the axon, our data suggest that Apaf1-deletion affects axonal outgrowth through an impairment of centrosome organization. In line with this, centrosomal protein expression, as well as their centrosomal localization proved to be altered upon Apaf1-deletion. Strikingly, we also found that Apaf1-loss affects trans-Golgi components and leads to a robust activation of AMP-dependent protein kinase (AMPK), this confirming the stressful conditions induced by Apaf1-deficiency. Since AMPK hyper-phosphorylation is known to impair a proper axon elongation, our finding contributes to explain the effect of Apaf1-deficiency on axogenesis. We also discovered that the signaling pathways mediating axonal growth and involving glycogen synthase kinase-3ß, liver kinase B1, and collapsing-response mediator protein-2 are altered in Apaf1-KO neurons. Overall, our results reveal a novel non-apoptotic role for Apaf1 in axonal outgrowth, suggesting that the neuronal phenotype due to Apaf1-deletion could not only be fully ascribed to apoptosis inhibition, but might also be the result of defects in axogenesis. The discovery of new molecules involved in axonal elongation has a clinical relevance since it might help to explain neurological abnormalities occurring during early brain development.


Subject(s)
Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Axons/pathology , Cerebral Cortex/pathology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Axons/physiology , Cell Differentiation , Centrosome/metabolism , Cerebral Cortex/embryology , Disks Large Homolog 4 Protein , Gene Expression Regulation, Developmental , Golgi Apparatus/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Neurons/cytology , Neurons/pathology , Neurons/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
6.
Nucleic Acids Res ; 40(9): 4086-96, 2012 May.
Article in English | MEDLINE | ID: mdl-22238374

ABSTRACT

The brain cytoplasmic RNA, BC1, is a small non-coding RNA that is found in different RNP particles, some of which are involved in translational control. One component of BC1-containing RNP complexes is the fragile X mental retardation protein (FMRP) that is implicated in translational repression. Peptide mapping and computational simulations show that the tudor domain of FMRP makes specific contacts to BC1 RNA. Endogenous BC1 RNA is 2'-O-methylated in nucleotides that contact the FMRP interface, and methylation can affect this interaction. In the cell body BC1 2'-O-methylations are present in both the nucleus and the cytoplasm, but they are virtually absent at synapses where the FMRP-BC1-mRNA complex exerts its function. These results strongly suggest that subcellular region-specific modifications of BC1 affect the binding to FMRP and the interaction with its mRNA targets. We finally show that BC1 RNA has an important role in translation of certain mRNAs associated to FMRP. All together these findings provide further insights into the translational regulation by the FMRP-BC1 complex at synapses.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Gene Expression Regulation , Protein Biosynthesis , RNA, Small Cytoplasmic/metabolism , Synapses/metabolism , Animals , Fragile X Mental Retardation Protein/chemistry , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Neurons/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA, Small Cytoplasmic/chemistry , RNA, Small Cytoplasmic/genetics
7.
Int J Mol Sci ; 15(4): 5596-622, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24694544

ABSTRACT

Apoptosis triggered by exogenous or endogenous stimuli is a crucial phenomenon to determine the fate of neurons, both in physiological and in pathological conditions. Our previous study established that gastric inhibitory polypeptide (Gip) is a neurotrophic factor capable of preventing apoptosis of cerebellar granule neurons (CGNs), during its pre-commitment phase. In the present study, we conducted whole-genome expression profiling to obtain a comprehensive view of the transcriptional program underlying the rescue effect of Gip in CGNs. By using DNA microarray technology, we identified 65 genes, we named survival related genes, whose expression is significantly de-regulated following Gip treatment. The expression levels of six transcripts were confirmed by real-time quantitative polymerase chain reaction. The proteins encoded by the survival related genes are functionally grouped in the following categories: signal transduction, transcription, cell cycle, chromatin remodeling, cell death, antioxidant activity, ubiquitination, metabolism and cytoskeletal organization. Our data outline that Gip supports CGNs rescue via a molecular framework, orchestrated by a wide spectrum of gene actors, which propagate survival signals and support neuronal viability.


Subject(s)
Apoptosis/genetics , Cerebellum/cytology , Gastric Inhibitory Polypeptide/metabolism , Neurons/cytology , Animals , Antioxidants , Cell Cycle Checkpoints/genetics , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Rats , Rats, Wistar , Signal Transduction , Transcription, Genetic , Ubiquitination/genetics
8.
J Neurosci ; 32(44): 15547-64, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23115191

ABSTRACT

A failure in the control of proliferation of cerebellar granule neuron precursor cells (GCPs), located in the external granular layer (EGL) of the cerebellum, gives rise to medulloblastoma. To investigate the process of neoplastic transformation of GCPs, we generated a new medulloblastoma model by crossing Patched1 heterozygous mice, which develop medulloblastomas with low frequency, with mice lacking the Tis21 gene. Overexpression of Tis21 is known to inhibit proliferation and trigger differentiation of GCPs; its expression decreases in human medulloblastomas. Double-knock-out mice show a striking increase in the frequency of medulloblastomas and hyperplastic EGL lesions, formed by preneoplastic GCPs. Tis21 deletion does not affect the proliferation of GCPs but inhibits their differentiation and, chiefly, their intrinsic ability to migrate outside the EGL. This defect of migration may represent an important step in medulloblastoma formation, as GCPs, remaining longer in the EGL proliferative niche, may become more prone to transformation. By genome-wide analysis, we identified the chemokine Cxcl3 as a target of Tis21. Cxcl3 is downregulated in Tis21-null GCPs of EGL and lesions; addition of Cxcl3 to cerebellar slices rescues the defective migration of Tis21-null GCPs and, remarkably, reduces the area of hyperplastic lesions. As Tis21 activates Cxcl3 transcription, our results suggest that Tis21 induces migration of GCPs through Cxcl3, which may represent a novel target for medulloblastoma therapy.


Subject(s)
Cell Movement/physiology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellum/cytology , Chemokines, CXC/physiology , Immediate-Early Proteins/genetics , Medulloblastoma/genetics , Neurons/physiology , Receptors, Cell Surface/genetics , Tumor Suppressor Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Bromodeoxyuridine , Cell Count , Cell Movement/genetics , Cell Proliferation , Chemokines, CXC/genetics , Genetic Vectors , Genotype , Heterozygote , Immediate-Early Proteins/physiology , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Medulloblastoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Patched Receptors , Patched-1 Receptor , Real-Time Polymerase Chain Reaction , Retroviridae/genetics , Tumor Suppressor Proteins/physiology
9.
Mol Cell Biochem ; 376(1-2): 189-95, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358924

ABSTRACT

Using in vitro models of Alzheimer's disease (AD), we found that the toxic effects of amyloid beta 25-35 (Aß(25-35)) on the neurotrophin brain-derived neurotrophic factor (BDNF) were counteracted by pre-incubation with neuropeptide Y (NPY), a neuropeptide expressed within the central nervous system. Nonetheless, the mechanism of action of NPY on BDNF neuronal production in the presence of Aß is not known. BDNF expression might be directly regulated by microRNA (miRs), small non-coding DNA fragments that regulate the expression of target genes. Thus, there is the possibility that miRs alterations are present in AD-affected neurons and that NPY influences miR expression. To test this hypothesis, we exposed NPY-pretreated primary rat cortical neurons to Aß(25-35) and measured miR-30a-5p (a member of the miR-30a family involved in BDNF tuning expression) and BDNF mRNA and protein expression after 24 and 48 h. Our results demonstrated that pre-treatment with NPY decreased miR-30a-5p expression and increased BDNF mRNA and protein expression at 24 and 48 h of incubation with Aß. Therefore, this study demonstrates that NPY modulates BDNF and its regulating microRNA miR-30a-5p in opposite direction with a mechanism that possibly contributes to the neuroprotective effect of NPY in rat cortical neurons exposed to Aß.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Brain-Derived Neurotrophic Factor/metabolism , MicroRNAs/metabolism , Neuropeptide Y/pharmacology , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Base Sequence , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/embryology , Gene Expression Regulation/drug effects , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/pharmacology , Rats , Rats, Wistar
10.
Cells ; 11(6)2022 03 20.
Article in English | MEDLINE | ID: mdl-35326503

ABSTRACT

RNA-binding proteins (RBPs) play important roles in modulating miRNA-mediated mRNA target repression. Argonaute2 (Ago2) is an essential component of the RNA-induced silencing complex (RISC) that plays a central role in silencing mechanisms via small non-coding RNA molecules known as siRNAs and miRNAs. Small RNAs loaded into Argonaute proteins catalyze endoribonucleolytic cleavage of target RNAs or recruit factors responsible for translational silencing and mRNA target destabilization. In previous studies we have shown that KCC2, a neuronal Cl (-) extruding K (+) Cl (-) co-transporter 2, is regulated by miR-92 in neuronal cells. Searching for Ago2 partners by immunoprecipitation and LC-MS/MS analysis, we isolated among other proteins the Serpine mRNA binding protein 1 (SERBP1) from SH-SY5Y neuroblastoma cells. Exploring the role of SERBP1 in miRNA-mediated gene silencing in SH-SY5Y cells and primary hippocampal neurons, we demonstrated that SERBP1 silencing regulates KCC2 expression through the 3' untranslated region (UTR). In addition, we found that SERBP1 as well as Ago2/miR-92 complex bind to KCC2 3'UTR. Finally, we demonstrated the attenuation of miR-92-mediated repression of KCC2 3'UTR by SERBP1 silencing. These findings advance our knowledge regarding the miR-92-mediated modulation of KCC2 translation in neuronal cells and highlight SERBP1 as a key component of this gene regulation.


Subject(s)
MicroRNAs , Symporters , 3' Untranslated Regions , Chromatography, Liquid , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA-Induced Silencing Complex/genetics , Symporters/genetics , Tandem Mass Spectrometry
11.
Proc Natl Acad Sci U S A ; 105(35): 13139-44, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18728191

ABSTRACT

Here, we report that interruption of NGF or BDNF signaling in hippocampal neurons rapidly activates the amyloidogenic pathway and causes neuronal apoptotic death. These events are associated with an early intracellular accumulation of PS1 N-terminal catalytic subunits and of APP C-terminal fragments and a progressive accumulation of intra- and extracellular Abeta aggregates partly released into the culture medium. The released pool of Abeta induces an increase of APP and PS1 holoprotein levels, creating a feed-forward toxic loop that might also cause the death of healthy neurons. These events are mimicked by exogenously added Abeta and are prevented by exposure to beta- and gamma-secretase inhibitors and by antibodies directed against Abeta peptides. The same cultured neurons deprived of serum die, but APP and PS1 overexpression does not occur, Abeta production is undetectable, and cell death is not inhibited by anti-Abeta antibodies, suggesting that hippocampal amyloidogenesis is not a simple consequence of an apoptotic trigger but is due to interruption of neurotrophic signaling.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/cytology , Nerve Growth Factor/metabolism , Neurons/metabolism , Signal Transduction , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/metabolism , Animals , Antibodies/pharmacology , Blotting, Western , Brain-Derived Neurotrophic Factor/pharmacology , Cell Death/drug effects , Cell Extracts , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Enzyme Inhibitors/pharmacology , Female , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/drug effects , Pregnancy , Rats , Rats, Wistar , Serum , Signal Transduction/drug effects
12.
Cells ; 10(11)2021 11 19.
Article in English | MEDLINE | ID: mdl-34831459

ABSTRACT

Neuronal apoptosis and survival are regulated at the transcriptional level. To identify key genes and upstream regulators primarily responsible for these processes, we overlayed the temporal transcriptome of cerebellar granule neurons following induction of apoptosis and their rescue by three different neurotrophic factors. We identified a core set of 175 genes showing opposite expression trends at the intersection of apoptosis and survival. Their functional annotations and expression signatures significantly correlated to neurological, psychiatric and oncological disorders. Transcription regulatory network analysis revealed the action of nine upstream transcription factors, converging pro-apoptosis and pro-survival-inducing signals in a highly interconnected functionally and temporally ordered manner. Five of these transcription factors are potential drug targets. Transcriptome-based computational drug repurposing produced a list of drug candidates that may revert the apoptotic core set signature. Besides elucidating early drivers of neuronal apoptosis and survival, our systems biology-based perspective paves the way to innovative pharmacology focused on upstream targets and regulatory networks.


Subject(s)
Apoptosis , Cell Lineage , Neurons/cytology , Transcription, Genetic , Animals , Apoptosis/genetics , Cell Survival/genetics , Cluster Analysis , Drug Repositioning , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Mental Disorders/genetics , Molecular Sequence Annotation , Nervous System Diseases/genetics , Neurons/metabolism , Protein Interaction Maps/genetics , Rats, Wistar , Time Factors , Transcription Factors/metabolism
13.
J Neurochem ; 113(3): 591-600, 2010 May.
Article in English | MEDLINE | ID: mdl-20050974

ABSTRACT

MicroRNAs have been associated to fine-tuning spatial and temporal control of gene expression during neuronal development. The neuronal Cl(-) extruding, K(+)Cl(-) co-transporter 2 (KCC2) is known to play an important role in neuronal Cl(-) homeostasis and in determining the physiological response to activation of anion selective GABA receptors. Here we show that microRNA-92 is developmentally down-regulated during maturation of rat cerebellar granule neurons (CGNs) in vitro. Computational predictions suggest several high-ranking targets for microRNA-92 including the KCC2 gene. Consistently, the KCC2 protein levels were up-regulated in mature CGN in vitro and a functional association between microRNA-92 and KCC2 3' untranslated region was established using luciferase assays. The generation of an inward directed Cl(-) electrochemical gradient, necessary for the hyperpolarizing effect of GABA, requires robust KCC2 expression in several neuronal types. Here we show that lentiviral-mediated microRNA-92 over-expression reduced KCC2 protein levels and positively shifted reversal potential of GABA induced Cl(-) currents in CGNs. In addition KCC2 re-expression reversed microRNA-92 electrophysiological phenotype. Consistently microRNA-92 inhibition induced both an increase of the level of KCC2 and a negative shift in GABA reversal potential. These findings introduce a new player in the developmental change of GABA from depolarization to hyperpolarization.


Subject(s)
Cerebellum/metabolism , MicroRNAs/pharmacology , Neurons/metabolism , Symporters/biosynthesis , 3' Untranslated Regions/genetics , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Cerebellum/cytology , Cerebellum/growth & development , Cytoplasmic Granules/metabolism , Electrophysiology , Gene Expression Regulation/physiology , Genes, Reporter/genetics , Genetic Vectors , Lentivirus/genetics , Luciferases/genetics , MicroRNAs/antagonists & inhibitors , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Wistar , Symporters/antagonists & inhibitors , gamma-Aminobutyric Acid/physiology , K Cl- Cotransporters
14.
Cells ; 9(12)2020 12 10.
Article in English | MEDLINE | ID: mdl-33321704

ABSTRACT

Neurotrophin nerve growth factor (NGF) has been demonstrated to upregulate the gene expression of bradykinin receptor 2 (B2R) on sensory neurons, thus facilitating nociceptive signals. The aim of the present study is to investigate the involvement of B2R in the NGF mechanism of action in nonsensory neurons in vitro by using rat mixed cortical primary cultures (CNs) and mouse hippocampal slices, and in vivo in Alzheimer's disease (AD) transgenic mice (5xFAD) chronically treated with NGF. A significant NGF-mediated upregulation of B2R was demonstrated by microarray, Western blot, and immunofluorescence analysis in CNs, indicating microglial cells as the target of this modulation. The B2R involvement in the NGF mechanism of action was also demonstrated by using a selective B2R antagonist which was able to reverse the neuroprotective effect of NGF in CNs, as revealed by viability assay, and the NGF-induced long-term potentiation (LTP) in hippocampal slices. To confirm in vitro observations, B2R upregulation was observed in 5xFAD mouse brain following chronic intranasal NGF treatment. This study demonstrates for the first time that B2R is a key element in the neuroprotective activity and synaptic plasticity mediated by NGF in brain cells.


Subject(s)
Alzheimer Disease/drug therapy , Nerve Growth Factor/administration & dosage , Neuroprotective Agents/administration & dosage , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Administration, Intranasal , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cell Survival , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Mice , Mice, Transgenic , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Nerve Growth Factor/pharmacology , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Primary Cell Culture , Rats , Up-Regulation
15.
Biomolecules ; 10(2)2020 02 01.
Article in English | MEDLINE | ID: mdl-32024191

ABSTRACT

In the last decade, Nerve Growth Factor (NGF)-based clinical approaches have lacked specific and efficient Tyrosine Kinase A (TrkA) agonists for brain delivery. Nowadays, the characterization of novel small peptidomimetic is taking centre stage in preclinical studies, in order to overcome the main size-related limitation in brain delivery of NGF holoprotein for Central Nervous System (CNS) pathologies. Here we investigated the NGF mimetic properties of the human NGF 1-14 sequence (hNGF1-14) and its derivatives, by resorting to primary cholinergic and dorsal root ganglia (DRG) neurons. Briefly, we observed that: 1) hNGF1-14 peptides engage the NGF pathway through TrkA phosphorylation at tyrosine 490 (Y490), and activation of ShcC/PI3K and Plc-γ/MAPK signalling, promoting AKT-dependent survival and CREB-driven neuronal activity, as seen by levels of the immediate early gene c-Fos, of the cholinergic marker Choline Acetyltransferase (ChAT), and of Brain Derived Neurotrophic Factor (BDNF); 2) their NGF mimetic activity is lost upon selective TrkA inhibition by means of GW441756; 3) hNGF1-14 peptides are able to sustain DRG survival and differentiation in absence of NGF. Furthermore, the acetylated derivative Ac-hNGF1-14 demonstrated an optimal NGF mimetic activity in both neuronal paradigms and an electrophysiological profile similar to NGF in cholinergic neurons. Cumulatively, the findings here reported pinpoint the hNGF1-14 peptide, and in particular its acetylated derivative, as novel, specific and low molecular weight TrkA specific agonists in both CNS and PNS primary neurons.


Subject(s)
Cholinergic Neurons/metabolism , Ganglia, Spinal/metabolism , Nerve Growth Factor/chemistry , Receptor, trkA/agonists , Receptor, trkA/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism , Animals , Biological Assay , Cell Differentiation , Cell Survival , Cells, Cultured , Humans , Peptides/chemistry , Phosphorylation , Rats , Signal Transduction , Tyrosine/chemistry
16.
J Neurochem ; 109(4): 1031-41, 2009 May.
Article in English | MEDLINE | ID: mdl-19519775

ABSTRACT

P2X receptors mediate a variety of physiological actions, including smooth muscle contraction, neuro-endocrine secretion and synaptic transmission. Among P2X receptors, the P2X(3) subtype is expressed in sensory neurons of dorsal root- and trigeminal-ganglia, where it performs a well-recognized role in sensory and pain transmission. Recent evidence indicates that the strength of P2X(3)-mediated responses is modulated in vivo by altering the number of receptors at the plasma membrane. In the present study, we investigate the trafficking properties of P2X(3) receptor in transfected HEK293 cells and in primary cultures of dorsal root ganglion neurons, finding that P2X(3) receptor undergoes rapid constitutive and cholesterol-dependent endocytosis. We also show that endocytosis is accompanied by preferential targeting of the receptor to late endosomes/lysosomes, with subsequent degradation. Furthermore, we observe that at steady state the receptor localizes predominantly in lamp1-positive intracellular structures, with a minor fraction present at the plasma membrane. Finally, the level of functional receptor expressed on the cell surface is rapidly up-regulated in response to agonist stimulation, which also augments receptor endocytosis. The findings presented in this work underscore a very dynamic trafficking behavior of P2X(3) receptor and disclose a possible mechanism for the rapid modulation of ATP-mediated responses potentially relevant during physiological and pathological conditions.


Subject(s)
Endocytosis/physiology , Purinergic P2 Receptor Agonists , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/physiology , Amino Acid Sequence , Animals , Biotinylation , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Electrophysiology , Endocytosis/drug effects , Endocytosis/genetics , Fluorescent Antibody Technique , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Humans , Ligands , Lysosomes/physiology , Molecular Sequence Data , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X3 , Signal Transduction/physiology , Transfection
17.
Mol Neurobiol ; 56(1): 535-552, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29736736

ABSTRACT

Basal forebrain cholinergic neurons (BFCN) are key modulators of learning and memory and are high energy-demanding neurons. Impaired neuronal metabolism and reduced insulin signaling, known as insulin resistance, has been reported in the early phase of Alzheimer's disease (AD), which has been suggested to be "Type 3 Diabetes." We hypothesized that BFCN may develop insulin resistance and their consequent failure represents one of the earliest event in AD. We found that a condition reminiscent of insulin resistance occurs in the medial septum of 3 months old 3×Tg-AD mice, reported to develop typical AD histopathology and cognitive deficits in adulthood. Further, we obtained insulin resistant BFCN by culturing them with high insulin concentrations. By means of these paradigms, we observed that nerve growth factor (NGF) reduces insulin resistance in vitro and in vivo. NGF activates the insulin receptor substrate 1 (IRS1) and rescues c-Fos expression and glucose metabolism. This effect involves binding of activated IRS1 to the NGF receptor TrkA, and is lost in presence of the specific IRS inhibitor NT157. Overall, our findings indicate that, in a well-established animal model of AD, the medial septum develops insulin resistance several months before it is detectable in the neocortex and hippocampus. Remarkably, NGF counteracts molecular alterations downstream of insulin-resistant receptor and its nasal administration restores insulin signaling in 3×Tg-AD mice by TrkA/IRS1 activation. The cross-talk between NGF and insulin pathways downstream the insulin receptor suggests novel potential therapeutic targets to slow cognitive decline in AD and diabetes-related brain insulin resistance.


Subject(s)
Alzheimer Disease/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Insulin/pharmacology , Nerve Growth Factor/pharmacology , Septal Nuclei/metabolism , Alzheimer Disease/genetics , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , Phosphorylation/drug effects , Rats , Rats, Wistar , Receptor, Insulin/metabolism , Septal Nuclei/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
19.
J Neurosci ; 27(33): 8885-92, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699670

ABSTRACT

Dopamine D(2) receptor (D(2)DR)-mediated transmission in the striatum is remarkably flexible, and changes in its efficacy have been heavily implicated in a variety of physiological and pathological conditions. Although receptor-associated proteins are clearly involved in specific forms of synaptic plasticity, the molecular mechanisms regulating the sensitivity of D(2) receptors in this brain area are essentially obscure. We have studied the physiological responses of the D(2)DR stimulations in mice lacking the brain cytoplasmic RNA BC1, a small noncoding dendritically localized RNA that is supposed to play a role in mRNA translation. We show that the efficiency of D(2)-mediated transmission regulating striatal GABA synapses is under the control of BC1 RNA, through a negative influence on D(2) receptor protein level affecting the functional pool of receptors. Ablation of the BC1 gene did not result in widespread dysregulation of synaptic transmission, because the sensitivity of cannabinoid CB(1) receptors was intact in the striatum of BC1 knock-out (KO) mice despite D(2) and CB(1) receptors mediated similar electrophysiological actions. Interestingly, the fragile X mental retardation protein FMRP, one of the multiple BC1 partners, is not involved in the BC1 effects on the D(2)-mediated transmission. Because D(2)DR mRNA is apparently equally translated in the BC1-KO and wild-type mice, whereas the protein level is higher in BC1-KO mice, we suggest that BC1 RNA controls D(2)DR indirectly, probably regulating translation of molecules involved in D(2)DR turnover and/or stability.


Subject(s)
Corpus Striatum/cytology , Neurons/physiology , Receptors, Dopamine D2/physiology , Ribonucleoproteins, Small Cytoplasmic/physiology , Synaptic Transmission/physiology , Animals , Animals, Newborn , Biphenyl Compounds/pharmacology , Cells, Cultured , Dopamine D2 Receptor Antagonists , Glutamate Decarboxylase/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Oligonucleotides/pharmacology , Patch-Clamp Techniques/methods , Piperazines/pharmacology , RNA, Long Noncoding , RNA, Messenger/biosynthesis , RNA, Untranslated , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/chemistry , Reverse Transcriptase Polymerase Chain Reaction/methods , Ribonucleoproteins, Small Cytoplasmic/deficiency , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
20.
J Neurochem ; 104(2): 534-44, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18173805

ABSTRACT

Different VGF peptides derived from Vgf, originally identified as a nerve growth factor responsive gene, have been detected in neurons within the central and peripheral nervous system and in various endocrine cells. In the current study, we have evaluated the ability of TLQP-21, a VGF-derived peptide, to protect, in a dose- and time-dependent manner, primary cultures of rat cerebellar granule cells (CGCs) from serum and potassium deprivation-induced cell death. We demonstrated that TLQP-21 increased survival of CGCs by decreasing the degree of apoptosis as assessed by cell viability and DNA fragmentation. Moreover, TLQP-21 significantly activated extracellular signal-regulated kinase 1/2, serine/threonine protein kinase, and c-jun N-terminal kinase phosphorylation, while decreased the extent of protein kinase C phosphorylation, as demonstrated by western blot analysis. In addition, TLQP-21 induced significant increase in intracellular calcium (as measured by fura-2AM) in about 60% of the recorded neurons. Taken together, the present results demonstrate that TLQP-21 promotes the survival of CGCs via pathways involving, within few minutes, modulation of kinases associated with CGCs survival, and by increasing intracellular calcium which can contribute to the neuroprotective effect of the peptide.


Subject(s)
Carrier Proteins/metabolism , Cerebellum/cytology , Neurons/drug effects , Peptide Fragments/pharmacology , Potassium/metabolism , Analysis of Variance , Animals , Butadienes/pharmacology , Calcium/metabolism , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/physiology , Nitriles/pharmacology , Phosphate-Binding Proteins , Potassium/pharmacology , Protein Kinases/metabolism , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL