Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
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
3.
Neuropharmacology ; 176: 108223, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32681844

ABSTRACT

Pro-nerve growth factor (proNGF) is the predominant form of NGF in the brain and its levels increase in neurodegenerative diseases. The balance between NGF receptors may explain the contradictory biological activities of proNGF. However, the specific role of the two main proNGF variants is mostly unexplored. proNGF-A is prevalently expressed in healthy brain, while proNGF-B content increases in the neuro-degenerating brain. Recently we have investigated in vitro the biological action of native mouse proNGF variants. To gain further insights into the specific functions of the two proNGFs, here we intranasally delivered mouse-derived proNGF-A and proNGF-B to the brain parenchyma of healthy and diabetic rats, the latter characterized by dysfunction in spatial learning and memory, in the septo-hippocampal circuitry and by relative increase in proNGF-B hippocampal levels. Exogenous proNGF-B induces depression of hippocampal DG-LTP and impairment of hippocampal neurogenesis in healthy animals, with concomitant decrease in basal forebrain cholinergic neurons and cholinergic fibers projecting to the hippocampus. proNGF-A, while ineffective in healthy animals, rescues the diabetes-induced impairment in DG-LTP and hippocampal neurogenesis, promoting the concomitant recovery of the basal forebrain cholinergic phenotype. Our experimental evidences suggest that the balance between different proNGFs may influence the development and progression of neurodegenerative diseases.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Drug Delivery Systems/methods , Hippocampus/metabolism , Nerve Growth Factor/administration & dosage , Nerve Net/metabolism , Protein Precursors/administration & dosage , Septum of Brain/metabolism , Administration, Intranasal , Animals , Female , Hippocampus/drug effects , Mice , Nerve Net/drug effects , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Septum of Brain/drug effects
4.
Nat Commun ; 10(1): 2572, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31189917

ABSTRACT

Activation of G-protein coupled receptors elevates cAMP levels promoting dissociation of protein kinase A (PKA) holoenzymes and release of catalytic subunits (PKAc). This results in PKAc-mediated phosphorylation of compartmentalized substrates that control central aspects of cell physiology. The mechanism of PKAc activation and signaling have been largely characterized. However, the modes of PKAc inactivation by regulated proteolysis were unknown. Here, we identify a regulatory mechanism that precisely tunes PKAc stability and downstream signaling. Following agonist stimulation, the recruitment of the chaperone-bound E3 ligase CHIP promotes ubiquitylation and proteolysis of PKAc, thus attenuating cAMP signaling. Genetic inactivation of CHIP or pharmacological inhibition of HSP70 enhances PKAc signaling and sustains hippocampal long-term potentiation. Interestingly, primary fibroblasts from autosomal recessive spinocerebellar ataxia 16 (SCAR16) patients carrying germline inactivating mutations of CHIP show a dramatic dysregulation of PKA signaling. This suggests the existence of a negative feedback mechanism for restricting hormonally controlled PKA activities.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Cyclic AMP/metabolism , Feedback, Physiological/physiology , Molecular Chaperones/metabolism , Spinocerebellar Ataxias/pathology , Animals , Feedback, Physiological/drug effects , Fibroblasts , HEK293 Cells , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Hippocampus/pathology , Holoenzymes/metabolism , Humans , Leupeptins/pharmacology , Mice , Mice, Inbred C57BL , Phosphorylation , Primary Cell Culture , Protein Binding/drug effects , Proteolysis/drug effects , Purine Nucleosides/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Spinocerebellar Ataxias/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology
5.
Front Neurosci ; 12: 154, 2018.
Article in English | MEDLINE | ID: mdl-29615849

ABSTRACT

We studied group-I metabotropic glutamate (mGlu) receptors in Pahenu2 (ENU2) mice, which mimic the genetics and neurobiology of human phenylketonuria (PKU), a metabolic disorder characterized, if untreated, by autism, and intellectual disability (ID). Male ENU2 mice showed increased mGlu5 receptor protein levels in the hippocampus and corpus striatum (but not in the prefrontal cortex) whereas the transcript of the mGlu5 receptor was unchanged. No changes in mGlu1 receptor mRNA and protein levels were found in any of the three brain regions of ENU2 mice. We extended the analysis to Homer proteins, which act as scaffolds by linking mGlu1 and mGlu5 receptors to effector proteins. Expression of the long isoforms of Homer was significantly reduced in the hippocampus of ENU2 mice, whereas levels of the short Homer isoform (Homer 1a) were unchanged. mGlu5 receptors were less associated to immunoprecipitated Homer in the hippocampus of ENU2 mice. The lack of mGlu5 receptor-mediated long-term depression (LTD) in wild-type mice (of BTBR strain) precluded the analysis of hippocampal synaptic plasticity in ENU2 mice. We therefore performed a behavioral analysis to examine whether pharmacological blockade of mGlu5 receptors could correct behavioral abnormalities in ENU2 mice. Using the same apparatus we sequentially assessed locomotor activity, object exploration, and spatial object recognition (spatial novelty test) after displacing some of the objects from their original position in the arena. Systemic treatment with the mGlu5 receptor antagonist, MPEP (20 mg/kg, i.p.), had a striking effect in the spatial novelty test by substantially increasing the time spent in exploring the displaced objects in ENU2 mice (but not in wild-type mice). These suggest a role for mGlu5 receptors in the pathophysiology of ID in PKU and suggest that, also in adult untreated animals, cognitive dysfunction may be improved by targeting these receptors with an appropriate therapy.

6.
Sci Rep ; 7(1): 9077, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831054

ABSTRACT

The muscarinic receptor response to acetylcholine regulates the hippocampal-related learning, memory, neural plasticity and the production and processing of the pro-nerve growth factor (proNGF) by hippocampal cells. The development and progression of diabetes generate a mild cognitive impairment reducing the functions of the septo-hippocampal cholinergic circuitry, depressing neural plasticity and inducing proNGF accumulation in the brain. Here we demonstrate, in a rat model of early type-1 diabetes, that a physical therapy, the electroacupuncture, counteracts the diabetes-induced deleterious effects on hippocampal physiology by ameliorating hippocampal-related memory functions; recovering the impaired long-term potentiation at the dentate gyrus (DG-LTP) and the lowered expression of the vesicular glutamate transporter 1; normalizing the activity-dependent release of proNGF in diabetic rat hippocampus. Electroacupuncture exerted its therapeutic effects by regulating the expression and activity of M1- and M2-acetylcholine muscarinic receptors subtypes in the dentate gyrus of hippocampus. Our results suggest that a physical therapy based on repetitive sensory stimulation could promote hippocampal neural activity, neuronal metabolism and functions, and conceivably improve the diabetes-induced cognitive impairment. Our data can support the setup of therapeutic protocols based on a better integration between physical therapies and pharmacology for the cure of diabetes-associated neurodegeneration and possibly for Alzheimer's disease.


Subject(s)
Electroacupuncture , Hippocampus/metabolism , Hippocampus/physiopathology , Muscarine/metabolism , Animals , Cell Count , Dentate Gyrus/metabolism , Dentate Gyrus/physiopathology , Diabetes Mellitus, Experimental , Long-Term Potentiation , Memory , Models, Biological , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neuronal Plasticity , Protein Precursors/genetics , Protein Precursors/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M2/metabolism , Receptors, Muscarinic/metabolism
7.
Synapse ; 71(5)2017 05.
Article in English | MEDLINE | ID: mdl-28187508

ABSTRACT

In the last two decades adiponectin, member of the adipokines family, gained attention because of its unique antidiabetic effects. However, the presence in the brain of adiponectin receptors and adiponectin itself raised interest because of the possible association with neuropsychiatric diseases. Indeed, clinical studies found altered concentration of adiponectin both in plasma and cerebrospinal fluid in several pathologies including depression, multiple sclerosis, Alzheimer's disease and stroke. Moreover, recent preclinical studies also suggest its involvement in different physiological functions. Despite this evidence very few studies attempted to elucidate the functional role of adiponectin at the synapse. To address this question, here we investigated the effect of Adiporon, an agonist of both adiponectin receptors on synaptic transmission and LTP at Schaffer-collateral CA1 pathway. Surprisingly, increasing concentration of Adiporon correlated with lower CA1-LTP levels and paired-pulse ratio, whereas basal transmission was always preserved. Collectively, our data show that the adiponectin system, beyond its involvement in metabolic diseases, plays also a critical role in synaptic activity thereby representing a putative target for the treatment of synaptic pathologies.


Subject(s)
CA1 Region, Hippocampal/metabolism , Receptors, Adiponectin/metabolism , Synaptic Transmission , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/physiology , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Receptors, Adiponectin/agonists
8.
Synapse ; 70(6): 223-30, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26850695

ABSTRACT

Homozygous or heterozygous mutations in the PTEN-induced kinase 1 (PINK1) gene have been linked to early-onset Parkinson's disease (PD). Several neurophysiological studies have demonstrated alterations in striatal synaptic plasticity along with impaired dopamine release in PINK1-deficient mice. Using electrophysiological methods, here we show that PINK1 loss of function causes a progressive increase of spontaneous glutamate-mediated synaptic events in the hippocampus, without influencing long-term potentiation. Moreover, fluorescence analysis reveals increased neurotrasmitter release although our biochemical results failed to detect which presynaptic proteins might be engaged. This study provides a novel role for PINK1 beyond the physiology of nigrostriatal dopaminergic circuit. Specifically, PINK1 might contribute to preserve synaptic function and glutamatergic homeostasis in the hippocampus, a brain region underlying cognition. The subtle changes in excitatory transmission here observed might be a pathogenic precursor to excitotoxic neurodegeneration and cognitive decline often observed in PD. Using electrophysiological and fluorescence techniques, we demonstrate that lack of PINK1 causes increased excitatory transmission and neurotransmitter release in the hippocampus, which might lead to the cognitive decline often observed in Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , Hippocampus/metabolism , Neuronal Plasticity/genetics , Parkinson Disease/metabolism , Protein Kinases/metabolism , Synaptic Transmission/genetics , Animals , Disease Models, Animal , Dopamine/metabolism , Mice, Knockout , Parkinson Disease/pathology , Protein Kinases/deficiency
9.
Mult Scler ; 22(11): 1405-1412, 2016 10.
Article in English | MEDLINE | ID: mdl-26733422

ABSTRACT

BACKGROUND: Alterations of synaptic transmission induced by inflammatory activity have been linked to the pathogenic mechanisms of multiple sclerosis (MS). Regulated upon activation, normal T-cell expressed, and secreted (RANTES) is a pro-inflammatory chemokine involved in MS pathophysiology, potentially able to regulate glutamate release and plasticity in MS brains, with relevant consequences on the clinical manifestations of the disease. OBJECTIVE: To assess the role of RANTES in the regulation of cortical excitability. METHODS: We explored the association of RANTES levels in the cerebrospinal fluid (CSF) of newly diagnosed MS patients with magnetic resonance imaging (MRI) and laboratory measures of inflammatory activity, as well its role in the control of cortical excitability and plasticity explored by means of transcranial magnetic stimulation (TMS), and in hippocampal mouse slices in vitro. RESULTS: CSF levels of RANTES were remarkably high only in active MS patients and were correlated with the concentrations of interleukin-1ß. RANTES levels were associated with TMS measures of cortical synaptic excitability, but not with long-term potentiation (LTP)-like plasticity. Similar findings were obtained in mouse hippocampal slices in vitro, where we observed that RANTES enhanced basal excitatory synaptic transmission with no effect on LTP. CONCLUSION: RANTES correlates with inflammation and synaptic excitability in MS brains.


Subject(s)
CA1 Region, Hippocampal/metabolism , Chemokine CCL5/cerebrospinal fluid , Cortical Excitability , Excitatory Postsynaptic Potentials/physiology , Multiple Sclerosis/cerebrospinal fluid , Adult , Animals , CA1 Region, Hippocampal/physiology , Case-Control Studies , Female , Hippocampus/metabolism , Hippocampus/physiology , Humans , In Vitro Techniques , Interleukin-1beta/cerebrospinal fluid , Long-Term Potentiation , Magnetic Resonance Imaging , Male , Mice , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/immunology , Transcranial Magnetic Stimulation
10.
Psychopharmacology (Berl) ; 233(4): 727-37, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26572895

ABSTRACT

RATIONALE: Compulsive symptoms develop in patients exposed to pramipexole (PPX), a dopaminergic agonist with high selectivity for the D3 receptor. Consistently, we demonstrated that PPX produces an exaggerated increase in contrafreeloading (CFL) for water, a repetitive and highly inflexible behavior that models core aspects of compulsive disorders. OBJECTIVES: Given the role of the hippocampus in behavioral flexibility, motivational control, and visuospatial working memory, we investigated the role of hippocampus in the expression of PPX-induced CFL. To this aim, rats were subjected to CFL under chronic PPX, and then examined for the electrophysiological, structural, and molecular properties of their hippocampus. METHODS: We measured long-term potentiation (LTP) at CA1 Schaffer collaterals, dendritic spine density in CA1 pyramidal neurons, and then glutamate release and expression of pre and postsynaptic proteins in hippocampal synaptosomes. The effects of PPX on hippocampal-dependent working memory were assessed through the novel object recognition (NOR) test. RESULTS: We found that PPX-treated rats showing CFL exhibited a significant decrease in hippocampal LTP and failed to exhibit the expected increase in hippocampal spine density. Glutamate release and PSD-95 expression were decreased, while pSYN expression was increased in hippocampal synaptosomes of PPX-treated rats showing CFL. Despite a general impairment of hippocampal synaptic function, working memory was unaffected by PPX treatment. CONCLUSIONS: Our findings demonstrate that chronic PPX affects synaptic function in the hippocampus, an area that is critically involved in the expression of flexible, goal-centered behaviors. We suggest that the hippocampus is a promising target in the pharmacotherapy of compulsive disorders.


Subject(s)
Benzothiazoles/administration & dosage , Dopamine Agonists/administration & dosage , Hippocampus/drug effects , Neural Inhibition/drug effects , Neuronal Plasticity/drug effects , Water/administration & dosage , Animals , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Male , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Pramipexole , Pyramidal Cells/drug effects , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism
11.
Proc Natl Acad Sci U S A ; 112(48): 14960-5, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26627246

ABSTRACT

Excitatory amino acids play a key role in both adaptive and deleterious effects of stressors on the brain, and dysregulated glutamate homeostasis has been associated with psychiatric and neurological disorders. Here, we elucidate mechanisms of epigenetic plasticity in the hippocampus in the interactions between a history of chronic stress and familiar and novel acute stressors that alter expression of anxiety- and depressive-like behaviors. We demonstrate that acute restraint and acute forced swim stressors induce differential effects on these behaviors in naive mice and in mice with a history of chronic-restraint stress (CRS). They reveal a key role for epigenetic up- and down-regulation of the putative presynaptic type 2 metabotropic glutamate (mGlu2) receptors and the postsynaptic NR1/NMDA receptors in the hippocampus and particularly in the dentate gyrus (DG), a region of active neurogenesis and a target of antidepressant treatment. We show changes in DG long-term potentiation (LTP) that parallel behavioral responses, with habituation to the same acute restraint stressor and sensitization to a novel forced-swim stressor. In WT mice after CRS and in unstressed mice with a BDNF loss-of-function allele (BDNF Val66Met), we show that the epigenetic activator of histone acetylation, P300, plays a pivotal role in the dynamic up- and down-regulation of mGlu2 in hippocampus via histone-3-lysine-27-acetylation (H3K27Ac) when acute stressors are applied. These hippocampal responses reveal a window of epigenetic plasticity that may be useful for treatment of disorders in which glutamatergic transmission is dysregulated.


Subject(s)
Behavior, Animal , Dentate Gyrus/metabolism , Epigenesis, Genetic , Glutamic Acid/metabolism , Stress, Psychological/metabolism , Synaptic Transmission , Animals , Anxiety/genetics , Anxiety/metabolism , Anxiety/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Dentate Gyrus/pathology , Depression/genetics , Depression/metabolism , Depression/pathology , Glutamic Acid/genetics , Histones/genetics , Histones/metabolism , Male , Mice , Mice, Mutant Strains , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Psychological/genetics , Stress, Psychological/pathology , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
12.
Front Cell Neurosci ; 9: 409, 2015.
Article in English | MEDLINE | ID: mdl-26528137

ABSTRACT

Adenosine is a widespread neuromodulator within the CNS and its extracellular level is increased during hypoxia or intense synaptic activity, modulating pre- and postsynaptic sites. We studied the neuromodulatory action of adenosine on glutamatergic currents in the hippocampus, showing that activation of multiple adenosine receptors (ARs) by basal adenosine impacts postsynaptic site. Specifically, the stimulation of both A1R and A3R reduces AMPA currents, while A2AR has an opposite potentiating effect. The effect of ARs stimulation on glutamatergic currents in hippocampal cultures was investigated using pharmacological and genetic approaches. A3R inhibition by MRS1523 increased GluR1-Ser845 phosphorylation and potentiated AMPA current amplitude, increasing the apparent affinity for the agonist. A similar effect was observed blocking A1R with DPCPX or by genetic deletion of either A3R or A1R. Conversely, impairment of A2AR reduced AMPA currents, and decreased agonist sensitivity. Consistently, in hippocampal slices, ARs activation by AR agonist NECA modulated glutamatergic current amplitude evoked by AMPA application or afferent fiber stimulation. Opposite effects of AR subtypes stimulation are likely associated to changes in GluR1 phosphorylation and represent a novel mechanism of physiological modulation of glutamatergic transmission by adenosine, likely acting in normal conditions in the brain, depending on the level of extracellular adenosine and the distribution of AR subtypes.

13.
Neurobiol Aging ; 36(5): 1890-902, 2015 May.
Article in English | MEDLINE | ID: mdl-25771393

ABSTRACT

We have investigated the relevance of d-aspartate oxidase, the only enzyme known to selectively degrade d-aspartate (d-Asp), in modulating glutamatergic system homeostasis. Interestingly, the lack of the Ddo gene, by raising d-Asp content, induces a substantial increase in extracellular glutamate (Glu) levels in Ddo-mutant brains. Consistent with an exaggerated and persistent N-methyl-d-aspartate receptor (NMDAR) stimulation, we documented in Ddo knockouts severe age-dependent structural and functional alterations mirrored by expression of active caspases 3 and 7 along with appearance of dystrophic microglia and reactive astrocytes. In addition, prolonged elevation of d-Asp triggered in mutants alterations of NMDAR-dependent synaptic plasticity associated to reduction of hippocampal GluN1 and GluN2B subunits selectively located at synaptic sites and to increase in the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-to-N-methyl-d-aspartate ratio. These effects, all of which converged on a progressive hyporesponsiveness at NMDAR sites, functionally resulted in a greater vulnerability to phencyclidine-induced prepulse inhibition deficits in mutants. In conclusion, our results indicate that d-aspartate oxidase, by strictly regulating d-Asp levels, impacts on the homeostasis of glutamatergic system, thus preventing accelerated neurodegenerative processes.


Subject(s)
D-Aspartate Oxidase/physiology , Glutamates/metabolism , Homeostasis/genetics , Mutation , Animals , Astrocytes/pathology , Caspase 3/metabolism , Caspase 7/metabolism , D-Aspartate Oxidase/genetics , D-Aspartic Acid/metabolism , Mice, Knockout , Microglia/pathology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/prevention & control , Neuronal Plasticity/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
14.
J Neurosci ; 34(13): 4558-66, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24672001

ABSTRACT

Angelman syndrome (AS) is caused by the loss of Ube3A, an ubiquitin ligase that commits specific proteins to proteasomal degradation. How this defect causes autism and other pathological phenotypes associated with AS is unknown. Long-term depression (LTD) of excitatory synaptic transmission mediated by type 5 metabotropic glutamate (mGlu5) receptors was enhanced in hippocampal slices of Ube3A(m-/p+) mice, which model AS. No changes were found in NMDA-dependent LTD induced by low-frequency stimulation. mGlu5 receptor-dependent LTD in AS mice was sensitive to the protein synthesis inhibitor anisomycin, and relied on the same signaling pathways as in wild-type mice, e.g., the mitogen-activated protein kinase (MAPK) pathway, the phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycine pathway, and protein tyrosine phosphatase. Neither the stimulation of MAPK and PI3K nor the increase in Arc (activity-regulated cytoskeleton-associated protein) levels in response to mGlu5 receptor activation were abnormal in hippocampal slices from AS mice compared with wild-type mice. mGlu5 receptor expression and mGlu1/5 receptor-mediated polyphosphoinositide hydrolysis were also unchanged in the hippocampus of AS mice. In contrast, AS mice showed a reduced expression of the short Homer protein isoform Homer 1a, and an increased coupling of mGlu5 receptors to Homer 1b/c proteins in the hippocampus. These findings support the link between Homer proteins and monogenic autism, and lay the groundwork for the use of mGlu5 receptor antagonists in AS.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/pathology , Carrier Proteins/metabolism , Hippocampus/physiopathology , Long-Term Synaptic Depression/physiology , Receptor, Metabotropic Glutamate 5/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Carrier Proteins/genetics , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hemizygote , Hippocampus/pathology , Homer Scaffolding Proteins , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism , Pyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology
15.
Biol Psychiatry ; 75(9): 701-10, 2014 May 01.
Article in English | MEDLINE | ID: mdl-23541633

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is characterized by the progressive degeneration of the nigrostriatal dopaminergic pathway and the emergence of rigidity, tremor, and bradykinesia. Accumulating evidence indicates that PD is also accompanied by nonmotor symptoms including cognitive deficits, often manifested as impaired visuospatial memory. METHODS: We studied cognitive performance and synaptic plasticity in a mouse model of PD, characterized by partial lesion of the dopaminergic and noradrenergic inputs to striatum and hippocampus. Sham- and 6-hydroxydopamine-lesioned mice were subjected to the novel object recognition test, and long-term potentiation was examined in the dentate gyrus and CA1 regions of the hippocampus. RESULTS: Bilateral 6-hydroxydopamine lesion reduced long-term but not short-term novel object recognition and decreased long-term potentiation specifically in the dentate gyrus. These abnormalities did not depend on the loss of noradrenaline but were abolished by the antiparkinsonian drug, L-DOPA, or by SKF81297, a dopamine D1-type receptor agonist. In contrast, activation of dopamine D2-type receptors did not modify the effects produced by the lesion. Blockade of the extracellular signal-regulated kinases prevented the ability of SKF81297 to rescue novel object recognition and long-term potentiation. CONCLUSIONS: These findings show that partial dopamine depletion leads to impairment of long-term recognition memory accompanied by abnormal synaptic plasticity in the dentate gyrus. They also demonstrate that activation of dopamine D1 receptors corrects these deficits, through a mechanism that requires intact extracellular signal-regulated kinases signaling.


Subject(s)
Cognition Disorders/physiopathology , Dentate Gyrus/physiopathology , Long-Term Potentiation/physiology , Parkinsonian Disorders/physiopathology , Recognition, Psychology/physiology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Dentate Gyrus/drug effects , Long-Term Potentiation/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice, Inbred C57BL , Neural Pathways/drug effects , Neural Pathways/physiopathology , Oxidopamine , Parkinsonian Disorders/complications , Parkinsonian Disorders/drug therapy , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Recognition, Psychology/drug effects , Synapses/drug effects , Synapses/physiology , Time Factors
16.
Neuromolecular Med ; 16(1): 38-51, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23892937

ABSTRACT

The immune system shapes synaptic transmission and plasticity in experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis (MS). These synaptic adaptations are believed to drive recovery of function after brain lesions, and also learning and memory deficits and excitotoxic neurodegeneration; whether inflammation influences synaptic plasticity in MS patients is less clear. In a cohort of 59 patients with MS, we found that continuous theta-burst transcranial magnetic stimulation did not induce the expected long-term depression (LTD)-like synaptic phenomenon, but caused persisting enhancement of brain cortical excitability. The amplitude of this long-term potentiation (LTP)-like synaptic phenomenon correlated with the concentration of the pro-inflammatory cytokine interleukin-1ß (IL-1ß) in the cerebrospinal fluid. In MS and EAE, the brain and spinal cord are typically enriched of CD3(+) T lymphocyte infiltrates, which are, along with activated microglia and astroglia, a major cause of inflammation. Here, we found a correlation between the presence of infiltrating T lymphocytes in the hippocampus of EAE mice and synaptic plasticity alterations. We observed that T lymphocytes from EAE, but not from control mice, release IL-1ß and promote LTP appearance over LTD, thereby mimicking the facilitated LTP induction observed in the cortex of MS patients. EAE-specific T lymphocytes were able to suppress GABAergic transmission in an IL-1ß-dependent manner, providing a possible synaptic mechanism able to lower the threshold of LTP induction in MS brains. Moreover, in vivo blockade of IL-1ß signaling resulted in inflammation and synaptopathy recovery in EAE hippocampus. These data provide novel insights into the pathophysiology of MS.


Subject(s)
Interleukin-1beta/physiology , Long-Term Potentiation/physiology , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Adolescent , Adult , Animals , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Gliosis/etiology , Gliosis/pathology , Hippocampus/physiopathology , Humans , Infusions, Intraventricular , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Random Allocation , T-Lymphocyte Subsets/metabolism , Theta Rhythm , Transcranial Magnetic Stimulation , Young Adult , gamma-Aminobutyric Acid/physiology
17.
J Neurosci ; 33(49): 19112-9, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24305808

ABSTRACT

Neuroplasticity is essential to prevent clinical worsening despite continuing neuronal loss in several brain diseases, including multiple sclerosis (MS). The precise nature of the adaptation mechanisms taking place in MS brains, ensuring protection from disability appearance and accumulation, is however unknown. Here, we explored the hypothesis that long-term synaptic potentiation (LTP), potentially able to minimize the effects of neuronal loss by providing extra excitation of denervated neurons, is the most relevant form of adaptive plasticity in stable MS patients, and it is disrupted in progressing MS patients. We found that LTP, explored by means of transcranial magnetic theta burst stimulation over the primary motor cortex, was still possible, and even favored, in stable relapsing-remitting (RR-MS) patients, whereas it was absent in individuals with primary progressive MS (PP-MS). We also provided evidence that platelet-derived growth factor (PDGF) plays a substantial role in favoring both LTP and brain reserve in MS patients, as this molecule: (1) was reduced in the CSF of PP-MS patients, (2) enhanced LTP emergence in hippocampal mouse brain slices, (3) was associated with more pronounced LTP in RR-MS patients, and (4) was associated with the clinical compensation of new brain lesion formation in RR-MS. Our results show that brain plasticity reserve, in the form of LTP, is crucial to contrast clinical deterioration in MS. Enhancing PDGF signaling might represent a valuable treatment option to maintain brain reserve and to attenuate the clinical consequences of neuronal damage in the progressive phases of MS and in other neurodegenerative disorders.


Subject(s)
Multiple Sclerosis/physiopathology , Neuronal Plasticity/physiology , Platelet-Derived Growth Factor/physiology , Signal Transduction/physiology , Synapses/physiology , Adult , Animals , Brain/physiology , Cerebral Cortex/physiology , Disease Progression , Electric Stimulation , Electrophysiological Phenomena , Evoked Potentials/physiology , Female , Humans , Long-Term Potentiation/physiology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis, Chronic Progressive/cerebrospinal fluid , Multiple Sclerosis, Chronic Progressive/physiopathology , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Platelet-Derived Growth Factor/cerebrospinal fluid , Theta Rhythm/physiology , Transcranial Magnetic Stimulation
18.
Drug Des Devel Ther ; 7: 1359-64, 2013.
Article in English | MEDLINE | ID: mdl-24255592

ABSTRACT

Alzheimer's disease, which is characterized by gradual cognitive decline associated with deterioration of daily living activities and behavioral disturbances throughout the course of the disease, is estimated to affect 27 million people around the world. It is expected that the illness will affect about 63 million people by 2030, and 114 million by 2050, worldwide. Current Alzheimer's disease medications may ease symptoms for a time but are not capable of slowing down disease progression. Indeed, all currently available therapies, such as cholinesterase inhibitors (donepezil, galantamine, rivastigmine), are primarily considered symptomatic therapies, although recent data also suggest possible disease-modifying effects. Gantenerumab is an investigational fully human anti-amyloid beta monoclonal antibody with a high capacity to bind and remove beta-amyloid plaques in the brain. This compound, currently undergoing Phase II and III clinical trials represents a promising agent with a disease-modifying potential in Alzheimer's disease. Here, we present an overview of gantenerumab ranging from preclinical studies to human clinical trials.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal/therapeutic use , Alzheimer Disease/physiopathology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Brain/drug effects , Brain/physiopathology , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Drug Evaluation, Preclinical , Humans
19.
Curr Pharm Des ; 19(36): 6480-90, 2013.
Article in English | MEDLINE | ID: mdl-23432715

ABSTRACT

The term "Autism Spectrum" is often used to describe disorders that are currently classified as Pervasive Developmental Disorders. These disorders are typically characterized by social deficits, communication difficulties, stereotyped or repetitive behaviors and/or cognitive delays or mental retardation; sometimes they present high comorbidity rates with epilepsy. Although these diagnoses share some common features, individuals with these disorders are thought to be "on the spectrum" because of differences in severity across these domains. Recent advances in the genetics of autism spectrum disorders (ASDs) are offering new valuable insights into molecular and cellular mechanisms of pathology. Of particular interest are transgenic technologies that allowed the engineering of several mouse models mimicking different kinds of monogenic heritable forms of ASDs. These transgenic models provide excellent opportunities to explore in detail cellular and molecular mechanisms underlying disease pathology and to identify novel targets for therapeutic intervention. Increasing evidence suggests that the pathophysiological core of the murine model is primarily due to changes in normal synaptic transmission and plasticity. Here, we will extensively review the synaptic alterations across different animal models of ASDs and recapitulate the pharmacological strategies aimed at rescuing hippocampal plasticity phenotypes. We describe how pharmacological modulation of mGlu5 receptor, through the use of positive or negative allosteric modulators (depending on the specific disorder), may represent a promising therapeutic strategy for ASDs treatment.


Subject(s)
Antipsychotic Agents/therapeutic use , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/therapy , Genetic Predisposition to Disease , Neuronal Plasticity/genetics , Synapses/drug effects , Allosteric Regulation/drug effects , Animals , Disease Models, Animal , Humans , Mice , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Synapses/pathology
20.
PLoS One ; 8(1): e54666, 2013.
Article in English | MEDLINE | ID: mdl-23355887

ABSTRACT

Abnormal use-dependent synaptic plasticity is universally accepted as the main physiological correlate of memory deficits in neurodegenerative disorders. It is unclear whether synaptic plasticity deficits take place during neuroinflammatory diseases, such as multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis (EAE). In EAE mice, we found significant alterations of synaptic plasticity rules in the hippocampus. When compared to control mice, in fact, hippocampal long-term potentiation (LTP) induction was favored over long-term depression (LTD) in EAE, as shown by a significant rightward shift in the frequency-synaptic response function. Notably, LTP induction was also enhanced in hippocampal slices from control mice following interleukin-1ß (IL-1ß) perfusion, and both EAE and IL-1ß inhibited GABAergic spontaneous inhibitory postsynaptic currents (sIPSC) without affecting glutamatergic transmission and AMPA/NMDA ratio. EAE was also associated with selective loss of GABAergic interneurons and with reduced gamma-frequency oscillations in the CA1 region of the hippocampus. Finally, we provided evidence that microglial activation in the EAE hippocampus was associated with IL-1ß expression, and hippocampal slices from control mice incubated with activated microglia displayed alterations of GABAergic transmission similar to those seen in EAE brains, through a mechanism dependent on enhanced IL-1ß signaling. These data may yield novel insights into the basis of cognitive deficits in EAE and possibly of MS.


Subject(s)
CA1 Region, Hippocampal , Encephalomyelitis, Autoimmune, Experimental , Long-Term Potentiation , Multiple Sclerosis , Synapses , Synaptic Transmission , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Cell Line , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Interleukin-1beta/metabolism , Mice , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , N-Methylaspartate/metabolism , Synapses/metabolism , Synapses/pathology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...