Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Mov Disord ; 38(2): 256-266, 2023 02.
Article in English | MEDLINE | ID: mdl-36350188

ABSTRACT

BACKGROUND: The accumulation of α-synuclein (α-syn) fibrils in intraneuronal inclusions called Lewy bodies and Lewy neurites is a pathological signature of Parkinson's disease (PD). Although several aspects linked to α-syn-dependent pathology (concerning its spreading, aggregation, and activation of inflammatory and neurodegenerative processes) have been under intense investigation, less attention has been devoted to the real impact of α-syn overexpression on structural and functional properties of substantia nigra pars compacta (SNpc) dopamine (DA) neurons, particularly at tardive stages of α-syn buildup, despite this has obvious relevance to comprehending mechanisms beyond PD progression. OBJECTIVES: We aimed to determine the consequences of a prolonged α-syn overexpression on somatodendritic morphology and functions of SNpc DA neurons. METHODS: We performed immunohistochemistry, stereological DA cell counts, analyses of dendritic arborization, ex vivo patch-clamp recordings, and in vivo DA microdialysis measurements in a 12- to 13-month-old transgenic rat model overexpressing the full-length human α-syn (Snca+/+ ) and age-matched wild-type rats. RESULTS: Aged Snca+/+ rats have mild loss of SNpc DA neurons and decreased basal DA levels in the SN. Residual nigral DA neurons display smaller soma and compromised dendritic arborization and, in parallel, increased firing activity, switch in firing mode, and hyperexcitability associated with hypofunction of fast activating/inactivating voltage-gated K+ channels and Ca2+ - and voltage-activated large conductance K+ channels. These intrinsic currents underlie the repolarization/afterhyperpolarization phase of action potentials, thus affecting neuronal excitability. CONCLUSIONS: Besides clarifying α-syn-induced pathological landmarks, such evidence reveals compensatory functional mechanisms that nigral DA neurons could adopt during PD progression to counteract neurodegeneration. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Parkinson Disease , Rats , Humans , Animals , Aged , Infant , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Dopaminergic Neurons/metabolism , Substantia Nigra/metabolism , Pars Compacta/metabolism , Rats, Transgenic
2.
Int J Mol Sci ; 23(17)2022 Sep 04.
Article in English | MEDLINE | ID: mdl-36077524

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by the accumulation of alpha-synuclein, encoded by the SNCA gene. The main neuropathological hallmark of PD is the degeneration of dopaminergic neurons leading to striatal dopamine depletion. Trophic support by a neurotrophin called glial-derived neurotrophic factor (GDNF) is also lacking in PD. We performed immunohistochemical studies to investigate neuropathological changes in the basal ganglia of a rat transgenic model of PD overexpressing alfa-synuclein. We observed that neuronal loss also occurs in the dorsolateral part of the striatum in the advanced stages of the disease. Moreover, along with the degeneration of the medium spiny projection neurons, we found a dramatic loss of parvalbumin interneurons. A marked decrease in GDNF, which is produced by parvalbumin interneurons, was observed in the striatum and in the substantia nigra of these animals. This confirmed the involvement of the striatum in the pathophysiology of PD and the importance of GDNF in maintaining the health of the substantia nigra.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Animals , Basal Ganglia/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Interneurons/metabolism , Parkinson Disease/genetics , Parvalbumins , Rats , Rats, Transgenic , Substantia Nigra/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
3.
Int J Mol Sci ; 23(9)2022 Apr 19.
Article in English | MEDLINE | ID: mdl-35562898

ABSTRACT

The degeneration of nigral dopaminergic neurons is considered the hallmark of Parkinson's disease (PD), and it is triggered by different factors, including mitochondrial dysfunction, Lewy body accumulation, neuroinflammation, excitotoxicity and metal accumulation. Despite the extensive literature devoted to unravelling the signalling pathways involved in neuronal degeneration, little is known about the functional impairments occurring in these cells during illness progression. Of course, it is not possible to obtain direct information on the properties of the dopaminergic cells in patients. However, several data are available in the literature reporting changes in the function of these cells in PD animal models. In the present manuscript, we focus on dopaminergic neuron functional properties and summarize shared or peculiar features of neuronal dysfunction in different PD animal models at different stages of the disease in an attempt to design a picture of the functional modifications occurring in nigral dopaminergic neurons during disease progression preceding their eventual death.


Subject(s)
Dopaminergic Neurons , Parkinson Disease , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Humans , Parkinson Disease/metabolism , Substantia Nigra/metabolism , alpha-Synuclein/metabolism
4.
Front Cell Neurosci ; 15: 798464, 2021.
Article in English | MEDLINE | ID: mdl-34924961

ABSTRACT

The cellular mechanisms regulating dopamine (DA) release in the striatum have attracted much interest in recent years. By in vitro amperometric recordings in mouse striatal slices, we show that a brief (5 min) exposure to the metabotropic glutamate receptor agonist DHPG (50 µM) induces a profound depression of synaptic DA release, lasting over 1 h from DHPG washout. This long-term depression is sensitive to glycine, which preferentially inhibits local cholinergic interneurons, as well as to drugs acting on nicotinic acetylcholine receptors and to the pharmacological depletion of released acetylcholine. The same DHPG treatment induces a parallel long-lasting enhancement in the tonic firing of presumed striatal cholinergic interneurons, measured with multi-electrode array recordings. When DHPG is bilaterally infused in vivo in the mouse striatum, treated mice display an anxiety-like behavior. Our results demonstrate that metabotropic glutamate receptors stimulation gives rise to a prolonged depression of the striatal dopaminergic transmission, through a sustained enhancement of released acetylcholine, due to the parallel long-lasting potentiation of striatal cholinergic interneurons firing. This plastic interplay between dopamine, acetylcholine, and glutamate in the dorsal striatum may be involved in anxiety-like behavior typical of several neuropsychiatric disorders.

5.
Mol Neurobiol ; 57(1): 586, 2020 01.
Article in English | MEDLINE | ID: mdl-31823196

ABSTRACT

The original version of this article unfortunately contained a mistake in Figure 3. The drawing superimposed on photomicrographs to identify the region of Dorsal raphè Nuclei was inappropriately positioned. The corrected figure is given below.

6.
ACS Med Chem Lett ; 10(4): 431-436, 2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30996775

ABSTRACT

The oxidative degeneration of dopamine-releasing (DAergic) neurons in the substantia nigra pars compacta (SNc) has attracted much interest in preclinical research, due to its involvement in Parkinson's disease manifestations. Evidence exists on the participation of quinone derivatives in mitochondrial dysfunction, alpha synuclein protein aggregation, and protein degradation. With the aim to investigate the role of L-DOPA-quinone in DAergic neuron functions, we synthesized L-DOPA-quinone by use of 2-iodoxybenzoic acid and measured its activity in recovery from dopamine-mediated firing inhibition of SNc neurons. Noteworthy, L-DOPA-quinone counteracts firing inhibition in SNc DAergic neurons caused by GIRK opening. A possible mechanism to explain the effect of L-DOPA-quinone on GIRK channel has been proposed by computational models. Overall, the study showed the possibility that L-DOPA-quinone stabilizes GIRK in a preopen conformation through formation of a covalent adduct with cysteine-65 on the GIRK2 subunit of the protein.

7.
Br J Pharmacol ; 175(17): 3534-3547, 2018 09.
Article in English | MEDLINE | ID: mdl-29933497

ABSTRACT

BACKGROUND AND PURPOSE: One of the hallmarks of ventral midbrain dopamine-releasing neurons is membrane hyperpolarization in response to stimulation of somato-dendritic D2 receptors. At early postnatal age, under sustained dopamine, this inhibitory response is followed by a slow recovery, resulting in dopamine inhibition reversal (DIR). In the present investigation, we aimed to get a better insight into the cellular mechanisms underlying DIR. EXPERIMENTAL APPROACH: We performed single-unit extracellular recordings with a multi-electrode array device and conventional patch-clamp recordings on midbrain mouse slices. KEY RESULTS: While continuous dopamine (100 µM) perfusion gave rise to firing inhibition that recovered in 10 to 15 min, the same effect was not obtained with the D2 receptor agonist quinpirole (100 nM). Moreover, firing inhibition caused by the GABAB receptor agonist baclofen (300 nM) was reversed by dopamine (100 µM), albeit D2 receptors had been blocked by sulpiride (10 µM). Conversely, the block of the dopamine transporter (DAT) with cocaine (30 µM) prevented firing recovery by dopamine under GABAB receptor stimulation. Accordingly, in whole-cell recordings from single cells, the baclofen-induced outward current was counteracted by dopamine (100 µM) in the presence of sulpiride (10 µM), and this effect was prevented by the DAT antagonists cocaine (30 µM) and GBR12909 (2 µM). CONCLUSIONS AND IMPLICATIONS: Our results indicate that the DAT plays a major role in DIR, mediating it under conditions of sustained dopamine exposure, and point to DAT as an important target for pharmacological therapies leading to prolonged enhancement of the dopaminergic signal.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/pharmacology , Substantia Nigra/drug effects , Animals , In Vitro Techniques , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Receptors, Dopamine D2/agonists , Substantia Nigra/cytology , Substantia Nigra/metabolism
8.
Neurobiol Dis ; 116: 142-154, 2018 08.
Article in English | MEDLINE | ID: mdl-29778899

ABSTRACT

The functional loop involving the ventral tegmental area (VTA), dorsal hippocampus and nucleus accumbens (NAc) plays a pivotal role in the formation of spatial memory and persistent memory traces. In particular, the dopaminergic innervation from the VTA to the hippocampus is critical for hippocampal-related memory function and alterations in the midbrain dopaminergic system are frequently reported in Alzheimer's disease (AD), contributing to age-related decline in memory and non-cognitive functions. However, much less is known about the hippocampus-NAc connectivity in AD. Here, we evaluated the functioning of the hippocampus-to-NAc core connectivity in the Tg2576 mouse model of AD that shows a selective and progressive degeneration of VTA dopaminergic neurons. We show that reduced dopaminergic innervation in the Tg2576 hippocampus results in reduced synaptic plasticity and excitability of dorsal subiculum pyramidal neurons. Importantly, the glutamatergic transmission from the hippocampus to the NAc core is also impaired. Chemogenetic depolarisation of Tg2576 subicular pyramidal neurons with an excitatory Designer Receptor Exclusively Activated by Designer Drugs, or systemic administration of the DA precursor levodopa, can both rescue the deficits in Tg2576 mice. Our data suggest that the dopaminergic signalling in the hippocampus is essential for the proper functioning of the hippocampus-NAc excitatory synaptic transmission.


Subject(s)
Alzheimer Disease/metabolism , Dopamine/metabolism , Hippocampus/metabolism , Nucleus Accumbens/metabolism , Synaptic Transmission/physiology , Alzheimer Disease/genetics , Animals , Dopamine/genetics , Dopaminergic Neurons/metabolism , Excitatory Postsynaptic Potentials/physiology , Male , Mice , Mice, Transgenic , Organ Culture Techniques
9.
Mol Neurobiol ; 55(10): 7921-7940, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29488136

ABSTRACT

Imbalances between excitatory and inhibitory synaptic transmission cause brain network dysfunction and are central to the pathogenesis of neurodevelopmental disorders. Parvalbumin interneurons are highly implicated in this imbalance. Here, we probed the social behavior and hippocampal function of mice carrying a haploinsufficiency for Ambra1, a pro-autophagic gene crucial for brain development. We show that heterozygous Ambra1 mice (Ambra+/-) are characterized by loss of hippocampal parvalbumin interneurons, decreases in the inhibition/excitation ratio, and altered social behaviors that are solely restricted to the female gender. Loss of parvalbumin interneurons in Ambra1+/- females is further linked to reductions of the inhibitory drive onto principal neurons and alterations in network oscillatory activity, CA1 synaptic plasticity, and pyramidal neuron spine density. Parvalbumin interneuron loss is underlined by increased apoptosis during the embryonic development of progenitor neurons in the medial ganglionic eminence. Together, these findings identify an Ambra1-dependent mechanism that drives inhibition/excitation imbalance in the hippocampus, contributing to abnormal brain activity reminiscent of neurodevelopmental disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Neural Inhibition , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/physiopathology , Animals , Apoptosis , Behavior, Animal , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Gamma Rhythm , Interneurons/metabolism , Male , Mice, Inbred C57BL , Neurodevelopmental Disorders/pathology , Neuronal Plasticity , Parvalbumins/metabolism , Social Behavior
10.
Mol Neurobiol ; 55(9): 7401-7412, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29417477

ABSTRACT

Recent studies show that microRNA-34 (miR-34) family is critical in the regulation of stress response also suggesting that it may contribute to the individual responsiveness to stress. We have recently demonstrated that mice carrying a genetic deletion of all miR-34 isoforms (triple knockout, TKO) lack the stress-induced serotonin (5-HT) and GABA release in the medial prefrontal cortex (mpFC) and basolateral amygdala (BLA), respectively. Here, we evaluated if the absence of miR-34 was also able to modify the stress-coping strategy in the forced swimming test. We found that the blunted neurochemical response to stress was associated with lower levels of immobility (index of active coping behavior) in TKO compared to WT mice. Interestingly, among the brain regions mostly involved in the stress-related behaviors, the miR-34 displayed the strongest expression in the dorsal raphe nuclei (DRN) of wild-type (WT) mice. In the DRN, the corticotropin-releasing factor receptors (CRFR) 1 and 2, contribute to determine the stress-coping style and the CRFR1 is a target of miR-34. Thus, we hypothesized that the miR-34-dependent modulation of CRFR1 expression may be involved in the DRN regulation of stress-coping strategies. In line with this hypothesis, we found increased CRFR1 levels in the DNR of TKO compared to WT mice. Moreover, infusion of CRFR1 antagonist in the DRN of TKO mice reverted their behavioral and neurochemical phenotype. We propose that miR-34 modulate the mpFC 5-HT/BLA GABA response to stress acting on CRFR1 in the DRN and that this mechanism could contribute to determine individual stress-coping strategy.


Subject(s)
Amygdala/metabolism , Behavior, Animal , MicroRNAs/metabolism , Prefrontal Cortex/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Serotonin/metabolism , Stress, Psychological/genetics , gamma-Aminobutyric Acid/metabolism , Acenaphthenes/pharmacology , Amygdala/drug effects , Animals , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/pharmacology , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Gene Deletion , Immobilization , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Motor Activity/drug effects , Swimming
11.
Nat Commun ; 8: 14727, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28367951

ABSTRACT

Alterations of the dopaminergic (DAergic) system are frequently reported in Alzheimer's disease (AD) patients and are commonly linked to cognitive and non-cognitive symptoms. However, the cause of DAergic system dysfunction in AD remains to be elucidated. We investigated alterations of the midbrain DAergic system in the Tg2576 mouse model of AD, overexpressing a mutated human amyloid precursor protein (APPswe). Here, we found an age-dependent DAergic neuron loss in the ventral tegmental area (VTA) at pre-plaque stages, although substantia nigra pars compacta (SNpc) DAergic neurons were intact. The selective VTA DAergic neuron degeneration results in lower DA outflow in the hippocampus and nucleus accumbens (NAc) shell. The progression of DAergic cell death correlates with impairments in CA1 synaptic plasticity, memory performance and food reward processing. We conclude that in this mouse model of AD, degeneration of VTA DAergic neurons at pre-plaque stages contributes to memory deficits and dysfunction of reward processing.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Dopaminergic Neurons/pathology , Memory , Reward , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Animals , Apoptosis/drug effects , Cell Death/drug effects , Dendritic Spines/metabolism , Dihydroxyphenylalanine/pharmacology , Dihydroxyphenylalanine/therapeutic use , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Food , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Inflammation/complications , Inflammation/pathology , Mice, Transgenic , Nerve Degeneration/complications , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Neuronal Plasticity/drug effects , Nucleus Accumbens/pathology , Nucleus Accumbens/physiopathology , Plaque, Amyloid/complications , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Selegiline/pharmacology , Selegiline/therapeutic use , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/pathology , Ventral Tegmental Area/physiopathology
12.
Front Mol Neurosci ; 10: 20, 2017.
Article in English | MEDLINE | ID: mdl-28232789

ABSTRACT

ProNGF, the precursor of mature Nerve Growth Factor (NGF), is the most abundant NGF form in the brain and increases markedly in the cortex in Alzheimer's Disease (AD), relative to mature NGF. A large body of evidence shows that the actions of ProNGF and mature NGF are often conflicting, depending on the receptors expressed in target cells. TgproNGF#3 mice, expressing furin-cleavage resistant proNGF in CNS neurons, directly reveal consequences of increased proNGF levels on brain homeostasis. Their phenotype clearly indicates that proNGF can be a driver of neurodegeneration, including severe learning and memory behavioral deficits, cholinergic deficits, and diffuse immunoreactivity for A-beta and A-beta-oligomers. In aged TgproNGF#3 mice spontaneous epileptic-like events are detected in entorhinal cortex-hippocampal slices, suggesting occurrence of excitatory/inhibitory (E/I) imbalance. In this paper, we investigate the molecular events linking increased proNGF levels to the epileptiform activity detected in hippocampal slices. The occurrence of spontaneous epileptiform discharges in the hippocampal network in TgproNGF#3 mice suggests an impaired inhibitory interneuron homeostasis. In the present study, we detect the onset of hippocampal epileptiform events at 1-month of age. Later, we observe a regional- and cellular-selective Parvalbumin interneuron and perineuronal net (PNN) depletion in the dentate gyrus (DG), but not in other hippocampal regions of TgproNGF#3 mice. These results demonstrate that, in the hippocampus, the DG is selectively vulnerable to altered proNGF/NGF signaling. Parvalbumin interneuron depletion is also observed in the amygdala, a region strongly connected to the hippocampus and likewise receiving cholinergic afferences. Transcriptome analysis of TgproNGF#3 hippocampus reveals a proNGF signature with broad down-regulation of transcription. The most affected mRNAs modulated at early times belong to synaptic transmission and plasticity and extracellular matrix (ECM) gene families. Moreover, alterations in the expression of selected BDNF splice variants were observed. Our results provide further mechanistic insights into the vicious negative cycle linking proNGF and neurodegeneration, confirming the regulation of E/I homeostasis as a crucial mediating mechanism.

13.
Eur J Neurosci ; 45(1): 92-105, 2017 01.
Article in English | MEDLINE | ID: mdl-27519559

ABSTRACT

We studied the properties of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA) in mice expressing the enhanced green fluorescent protein (eGFP) under the control of the tyrosine hydroxylase promoter (TH-GFP). By using a practical map of cell positioning in distinct SNpc and VTA subregions in horizontal midbrain slices we saw that the spontaneous firing, membrane properties, cell body size and magnitude of the hyperpolarization-activated current (Ih ) in TH-GFP-positive neurons (TH-GFP+ ) vary significantly among subregions, following a mediolateral gradient. Block of Ih with Zd7288 inhibited firing in the most lateral subregions, but had little effect in the intermediate/medial VTA. In addition, TH-GFP+ cells were excited by Met5 -Enkephalin. Extracellular recordings from a large neuron number showed that all TH-GFP+ cells were inhibited by dopamine, suggesting that this is a reliable approach for identifying dopaminergic neurons in vitro. Simultaneous recordings from dopamine-sensitive and dopamine-insensitive neurons showed that dopamine-insensitive cells (putative non-dopaminergic neurons) are unaffected by Zd7288 but inhibited by Met5 -Enkephalin. Under patch-clamp, dopamine generated a quantitatively similar outward current in most TH-GFP+ neurons, although medial VTA cells showed reduced dopamine sensitivity. Pargyline prolonged the dopamine current, whereas cocaine enhanced dopamine-mediated responses in both the SNpc and the VTA. Our work provides new insights into the variability in mouse midbrain dopaminergic neurons along the medial-lateral axis and points to the necessity of a combination of different electrophysiological and pharmacological approaches for reliably identifying these cells to distinguish them from non-dopaminergic neurons in the midbrain.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Substantia Nigra/metabolism , Ventral Tegmental Area/metabolism , Animals , Cells, Cultured , Female , Male , Membrane Potentials/physiology , Mesencephalon/metabolism , Mice , Tyrosine 3-Monooxygenase/metabolism
14.
Exp Neurol ; 287(Pt 1): 21-33, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27771352

ABSTRACT

The presence of α-synuclein (α-syn) in Lewy bodies and Lewy neurites is an important characteristic of the neurodegenerative processes of substantia nigra pars compacta (SNpc) dopaminergic (DAergic) neurons in Parkinson's disease (PD) and other synucleinopathies. Here we report that Berlin-Druckrey rats carrying a spontaneous mutation in the 3' untranslated region of α-syn mRNA (m/m rats) display a marked accumulation of α-syn in the mesencephalic area, striatum and frontal cortex, accompanied to severe dysfunctions in the dorsolateral striatum. Despite a small reduction in the number of SNpc and ventral tegmental area DAergic cells, the surviving dopaminergic neurons of the m/m rats do not show clear-cut alterations of the spontaneous and evoked firing activity, DA responses and somatic amphetamine-induced firing inhibition. Interestingly, mutant DAergic neurons display diminished whole-cell Ih conductance and a reduced frequency of spontaneous excitatory synaptic currents. By contrast, m/m rats show a severe impairment of DA and glutamate release in the dorsolateral striatum, as revealed by amperometric measure of DA currents and by electrophysiological recordings of glutamatergic synaptic events in striatal medium spiny neurons. These functional impairments are paralleled by a decreased expression of the DA transporter and VGluT1 proteins in the same area. Thus, together with α-syn overload in the mesencephalic region, striatum and frontal cortex, the main functional alterations occur in the DAergic and glutamatergic terminals in the dorsal striatum of the m/m rats.


Subject(s)
Dopaminergic Neurons/physiology , Glutamic Acid/metabolism , Membrane Potentials/physiology , Mesencephalon/cytology , alpha-Synuclein/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Bicuculline/pharmacology , Cell Count , Dopamine/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , GABA-B Receptor Agonists/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Patch-Clamp Techniques , Picrotoxin/pharmacology , Rats , Synaptic Potentials/drug effects , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , alpha-Synuclein/genetics
15.
Neuromolecular Med ; 18(1): 50-68, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26530396

ABSTRACT

Transgenic mice overexpressing spermine oxidase (SMO) in the cerebral cortex (Dach-SMO mice) showed increased vulnerability to excitotoxic brain injury and kainate-induced epileptic seizures. To investigate the mechanisms by which SMO overexpression leads to increased susceptibility to kainate excitotoxicity and seizure, in the cerebral cortex of Dach-SMO and control mice we assessed markers for astrocyte proliferation and neuron loss, and the ability of kainate to evoke glutamate release from nerve terminals and astrocyte processes. Moreover, we assessed a possible role of astrocytes in an in vitro model of epileptic-like activity in combined cortico-hippocampal slices recorded with a multi-electrode array device. In parallel, as the brain is a major metabolizer of oxygen and yet has relatively feeble protective antioxidant mechanisms, we analyzed the oxidative status of the cerebral cortex of both SMO-overexpressing and control mice by evaluating enzymatic and non-enzymatic scavengers such as metallothioneins. The main findings in the cerebral cortex of Dach-SMO mice as compared to controls are the following: astrocyte activation and neuron loss; increased oxidative stress and activation of defense mechanisms involving both neurons and astrocytes; increased susceptibility to kainate-evoked cortical epileptogenic activity, dependent on astrocyte function; appearance of a glutamate-releasing response to kainate from astrocyte processes due to activation of Ca(2+)-permeable AMPA receptors in Dach-SMO mice. We conclude that reactive astrocytosis and activation of glutamate release from astrocyte processes might contribute, together with increased reactive oxygen species production, to the vulnerability to kainate excitotoxicity in Dach-SMO mice. This mouse model with a deregulated polyamine metabolism would shed light on roles for astrocytes in increasing vulnerability to excitotoxic neuron injury.


Subject(s)
Astrocytes/drug effects , Kainic Acid/pharmacology , Nerve Tissue Proteins/physiology , Neurotoxins/pharmacology , Oxidoreductases Acting on CH-NH Group Donors/physiology , Seizures/chemically induced , Animals , Aspartic Acid/metabolism , Astrocytes/pathology , Benzodiazepines/pharmacology , Biogenic Polyamines/metabolism , Calcium/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Enzyme Induction , Genetic Predisposition to Disease , Gliosis/genetics , Hippocampus/enzymology , Hippocampus/pathology , Hippocampus/physiopathology , Male , Metallothionein/physiology , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/pathology , Oxidative Stress , Oxidoreductases Acting on CH-NH Group Donors/biosynthesis , Oxidoreductases Acting on CH-NH Group Donors/genetics , Receptors, AMPA/drug effects , Receptors, AMPA/physiology , Recombinant Fusion Proteins/biosynthesis , Seizures/genetics , Seizures/physiopathology , Synaptosomes/drug effects , Synaptosomes/physiology , Up-Regulation , Polyamine Oxidase
16.
Exp Neurol ; 261: 733-43, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25173217

ABSTRACT

γ-Aminobutyric acid A receptor (GABAAR)-mediated postsynaptic currents were recorded in brain slices from substantia nigra pars reticulate neurons. The selective adenosine A1 receptor (A1R) antagonist, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), increased the frequency, but not the amplitude of spontaneous inhibitory post-synaptic currents (IPSCs) in the presence of the dopamine D1 receptor agonist SKF 38393 (SKF) and phosphodiesterase 10A inhibitors (papaverine or AE90074). Under these conditions, DPCPX also increased the amplitude of evoked IPSCs (eIPSCs). The effect of DPCPX was also examined in a mouse model of Parkinson's disease (PD), generated by unilateral denervation of the dopaminergic input to the striatum. In this model, SKF alone was sufficient to increase sIPSCs frequency and eIPSCs amplitude, and these effects were not potentiated by DPCPX. To confirm a depressive effect of A1Rs on the synaptic release of GABA we used the selective A1R agonist 5'-chloro-5'-deoxy-N(6)-(±)-(endo-norborn-2-yl)adenosine (5'Cl5'd-(±)-ENBA) which has limited peripheral actions. We found that 5'Cl5'd-(±)-ENBA decreased sIPSCs frequency, without affecting their amplitude, and decreased eIPSCs amplitude. Importantly, in the PD mouse model, 5'Cl5'd-(±)-ENBA prevented the increase in sIPSC frequency and eIPSC amplitude produced by SKF. Since exaggerated DA transmission along the striato-nigral pathway is involved in the motor complications (e.g. dyskinesia) caused by prolonged and intermittent administration of l-DOPA, we examined the effect of A1R activation in mice with unilateral DA denervation. We found that 5'Cl5'd-(±)-ENBA, administered in combination with l-DOPA, reduced the development of abnormal involuntary movements. These results indicate the potential benefit of A1R agonists for the treatment of l-DOPA-induced dyskinesia and hyperkinetic disorders providing a mechanistic framework for the study of the interaction between DA and adenosine in the striatonigral system.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Dyskinesia, Drug-Induced/drug therapy , Receptor, Adenosine A1/metabolism , Receptors, Dopamine D1/metabolism , Action Potentials/drug effects , Age Factors , Animals , Antiparkinson Agents/adverse effects , Corpus Striatum/drug effects , Disease Models, Animal , Dyskinesia, Drug-Induced/etiology , Enzyme Inhibitors/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Levodopa/adverse effects , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurons/drug effects , Neurons/physiology , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Pars Reticulata/cytology , Pars Reticulata/drug effects , Xanthines/pharmacology
17.
Neuropharmacology ; 76 Pt A: 127-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23973317

ABSTRACT

In the present study, we found that PDE10A inhibitor papaverine, alone or in combination with the D1 receptor agonist SKF38393, did not change spontaneous IPSCs (sIPSCs) frequency or amplitude in the substantia nigra pars reticulata (SNpr). An increase in frequency, but not in amplitude, of sIPSCs was only observed when SKF38393 and PDE10A inhibitors were associated to perfusion with higher extracellular K(+). On the other hand, the amplitude of evoked IPSCs (eIPSCs) of the striato-nigral projection to SNpr, was increased in response to co-administration of SKF38393 and papaverine in normal extracellular potassium. Of note, both an increase in sIPSCs frequency and eIPSC amplitude could be obtained either by a robust stimulation of adenylyl cyclase (AC) with forskolin (10 µM) or by a lower dose of forskolin (1 µM) associated to PDE inhibition. We next investigated the effects produced by dopamine (DA) depletion in the striatum. Under this condition, SKF38393 alone increased either sIPSCs frequency and eIPSC amplitude. In addition, in the striatum of DA-depleted mice we found reduced PDE10A levels and higher cAMP-dependent phosphorylation in response to D1 receptor stimulation. In accordance with these biochemical data, perfusion with papaverine had no effect on the SKF38393-induced changes of IPSCs in slices of DA-depleted mice. These findings reveal a dynamic interplay between PDE10A activity, level of neuronal network depolarization and degree of dopaminergic tone in the ability of D1 receptors to facilitate the GABAergic transmission to SNpr neurons from the direct nigro-striatal pathway. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.


Subject(s)
Corpus Striatum/physiology , Dopamine/deficiency , Dopamine/metabolism , Neural Pathways/physiology , Phosphoric Diester Hydrolases/physiology , Receptors, Dopamine D1/metabolism , Substantia Nigra/physiology , gamma-Aminobutyric Acid/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Colforsin/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Dopamine Agonists/pharmacology , Drug Interactions , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Mice , Neural Pathways/drug effects , Oxidopamine/pharmacology , Papaverine/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Phosphorylation/drug effects , Potassium/pharmacology , Substantia Nigra/drug effects , Substantia Nigra/enzymology
18.
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
19.
Neuromolecular Med ; 15(3): 541-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23821337

ABSTRACT

Functional and ultrastructural investigations support the concept that altered brain connectivity, exhausted neural plasticity, and synaptic loss are the strongest correlates of cognitive decline in age-related neurodegenerative dementia of Alzheimer's type. We have previously demonstrated that in transgenic mice, expressing amyloid-ß precursor protein-Swedish mutation active caspase-3 accumulates in hippocampal postsynaptic compartments leading to altered postsynaptic density (PSD) composition, increased long-term depression (LTD), and dendritic spine loss. Furthermore, we found strong evidence that dendritic spine alteration is mediated by calcineurin activation, a calcium-dependent phosphatase involved in synapse signaling. In the present work, we analyzed the molecular mechanism linking alteration of synaptic plasticity to the increase of calcineurin activity. We found that acute treatment of young and plaque-free transgenic mice with the calcineurin inhibitor FK506 leads to a complete rescue of LTD and PSD composition. Our findings are in agreement with other results reporting that calcineurin inhibition improves memory function and restores dendritic spine density, confirming that calcineurin inhibition may be explored as a neuroprotective treatment to stop or slowdown synaptic alterations in Alzheimer's disease.


Subject(s)
Alzheimer Disease/prevention & control , CA1 Region, Hippocampal/drug effects , Calcineurin Inhibitors , Long-Term Synaptic Depression/drug effects , Neuroprotective Agents/therapeutic use , Post-Synaptic Density/drug effects , Tacrolimus/therapeutic use , Alzheimer Disease/physiopathology , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Caspase 3/metabolism , Dendrites/drug effects , Dendrites/ultrastructure , Disease Models, Animal , Disks Large Homolog 4 Protein , Drug Evaluation, Preclinical , Excitatory Postsynaptic Potentials/drug effects , Guanylate Kinases/biosynthesis , Guanylate Kinases/genetics , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Transgenic , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Processing, Post-Translational/drug effects , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/agonists , Tacrolimus/pharmacology
20.
Exp Neurol ; 247: 582-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23481547

ABSTRACT

L-DOPA (Levodopa) remains the gold standard for the treatment of motor symptoms of Parkinson's disease (PD), despite indications that the drug may have detrimental effects in cell culture. Classically, l-DOPA increases the production of dopamine (DA) in nigral dopaminergic neurons, while paradoxically inhibiting the firing of these neurons due to activation of D2 autoreceptors by extracellularly released DA. Using a combination of electrophysiology and calcium microfluorometry in brain slices, we have identified a novel effect of L-DOPA on dopaminergic neurons when D2 receptors were blocked. Under these conditions, L-DOPA (0.03-3 mM) evoked an excitatory effect consisting of two components. The 'early' component observed during and immediately after application of the drug, was associated with increased firing, membrane depolarization and inward current. This excitatory response was strongly attenuated by CNQX (10 µM), pointing to the involvement of TOPA quinone, an auto-oxidation product of L-DOPA and a potent activator of AMPA/kainate receptors. The 'late' phase of excitation persisted >30 min after brief L-DOPA application and was not mediated by ionotropic glutamate receptors, nor by D1, α1-adrenergic, mGluR1 or GABAB receptors. It was eliminated by carbidopa, demonstrating its dependence on conversion of L-DOPA to DA. Exogenous DA (50 µM) also evoked a glutamate-receptor independent increase in firing and an inward current when D2 receptors were blocked. In voltage-clamped neurons, both L-DOPA and DA produced a long-lasting increase in [Ca(2+)]i which was unaffected by block of ionotropic glutamate receptors. These results demonstrate that L-DOPA has dual, inhibitory and excitatory, effects on nigral dopaminergic neurons, and suggest that the excitation and calcium rise may have long-lasting consequences for the activity and survival of these neurons when the expression or function of D2 receptors is impaired.


Subject(s)
Dopamine Agents/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Levodopa/pharmacology , Substantia Nigra/cytology , Action Potentials/drug effects , Analysis of Variance , Animals , Animals, Newborn , Calcium/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , In Vitro Techniques , Mice , Patch-Clamp Techniques , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...