Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.786
Filtrer
1.
Behav Neurosci ; 138(3): 164-177, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38934920

RÉSUMÉ

A growing body of literature indicates that mediated learning techniques have specific utility for tapping into reality testing in animal models of neuropsychiatric illness. In particular, recent work has shown that animal models that recapitulate various endophenotypes of schizophrenia are particularly vulnerable to impairments in reality testing when undergoing mediated learning. Multiple studies have indicated that these effects are dopamine receptor 2-dependent and correlated with aberrant insular cortex (IC) activity. However, until now, the connection between dopamine and the IC had not been investigated. Here, we utilized a novel intersectional approach to label mesencephalic dopamine cells that specifically project to the insular cortex in both wild-type controls and transgenic mice expressing the dominant-negative form of the Disrupted-in-Schizophrenia-1 (DISC-1) gene. Using these techniques, we identified a population of cells that project from the ventral tegmental area (VTA) to the IC. Afterward, we conducted multiple studies to test the necessity of this circuit in behaviors ranging from gustatory detection to the maintenance of effort and, finally, mediated performance. Our results indicate that perturbations of the DISC-1 genetic locus lead to a reduction in the number of cells in the VTA → IC circuit. Behaviorally, VTA → IC circuitry does not influence gustatory detection or motivation to acquire sucrose reward; however, inactivation of this circuit differentially suppresses Pavlovian approach behavior in wild-type and DISC-1 transgenic mice during mediated performance testing. Moreover, under these testing conditions, inactivation of this circuit predisposes wild-type (but not DISC-1) mice to display impaired reality testing. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Sujet(s)
Neurones dopaminergiques , Cortex insulaire , Souris transgéniques , Animaux , Neurones dopaminergiques/physiologie , Neurones dopaminergiques/métabolisme , Souris , Cortex insulaire/physiologie , Mâle , Aire tegmentale ventrale/physiologie , Aire tegmentale ventrale/métabolisme , Souris de lignée C57BL , Voies nerveuses/physiologie , Récompense , Modèles animaux de maladie humaine , Dopamine/métabolisme , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Mésencéphale/métabolisme , Mésencéphale/physiologie , Schizophrénie/physiopathologie
2.
Cells ; 13(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38891076

RÉSUMÉ

Pacemaking activity in substantia nigra dopaminergic neurons is generated by the coordinated activity of a variety of distinct somatodendritic voltage- and calcium-gated ion channels. We investigated whether these functional interactions could arise from a common localization in macromolecular complexes where physical proximity would allow for efficient interaction and co-regulations. For that purpose, we immunopurified six ion channel proteins involved in substantia nigra neuron autonomous firing to identify their molecular interactions. The ion channels chosen as bait were Cav1.2, Cav1.3, HCN2, HCN4, Kv4.3, and SK3 channel proteins, and the methods chosen to determine interactions were co-immunoprecipitation analyzed through immunoblot and mass spectrometry as well as proximity ligation assay. A macromolecular complex composed of Cav1.3, HCN, and SK3 channels was unraveled. In addition, novel potential interactions between SK3 channels and sclerosis tuberous complex (Tsc) proteins, inhibitors of mTOR, and between HCN4 channels and the pro-degenerative protein Sarm1 were uncovered. In order to demonstrate the presence of these molecular interactions in situ, we used proximity ligation assay (PLA) imaging on midbrain slices containing the substantia nigra, and we could ascertain the presence of these protein complexes specifically in substantia nigra dopaminergic neurons. Based on the complementary functional role of the ion channels in the macromolecular complex identified, these results suggest that such tight interactions could partly underly the robustness of pacemaking in dopaminergic neurons.


Sujet(s)
Neurones dopaminergiques , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation , Mésencéphale , Protéomique , Canaux potassiques calcium-dépendants de petite conductance , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation/métabolisme , Protéomique/méthodes , Neurones dopaminergiques/métabolisme , Animaux , Canaux potassiques calcium-dépendants de petite conductance/métabolisme , Mésencéphale/métabolisme , Humains , Canaux calciques de type L/métabolisme , Souris , Substantia nigra/métabolisme
3.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891832

RÉSUMÉ

The loss of midbrain dopaminergic (DA) neurons is the fundamental pathological feature of Parkinson's disease (PD). PD causes chronic pain in two-thirds of patients. Recent studies showed that the activation of the pedunculopontine tegmental nucleus (PPTg) can effectively relieve inflammatory pain and neuropathic pain. The PPTg is located in the pontomesencephalic tegmentum, a target of deep brain stimulation (DBS) treatment in PD, and is involved in motor control and sensory integration. To test whether the lesion of midbrain DA neurons induced pain hypersensitivity, and whether the chemogenetic activation of the PPTg could modulate the pain, the AAV-hM3Dq receptor was transfected and expressed into the PPTg neurons of 6-hydroxydopamine-lesioned mice. In this study, von Frey, open field, and adhesive tape removal tests were used to assess animals' pain sensitivity, locomotor activity, and sensorimotor function and somatosensory perception, respectively. Here, we found that the lesion of midbrain DA neurons induced a minor deficit in voluntary movement but did not affect sensorimotor function and somatosensory perception in the tape removal test. The results showed that lesion led to pain hypersensitivity, which could be alleviated both by levodopa and by the chemogenetic activation of the PPTg. Activating the PPTg may be a potential therapeutic strategy to relieve pain phenotypes in PD.


Sujet(s)
Neurones dopaminergiques , Mésencéphale , Noyau tegmental pédonculopontin , Animaux , Noyau tegmental pédonculopontin/métabolisme , Neurones dopaminergiques/métabolisme , Souris , Mésencéphale/métabolisme , Mâle , Maladie de Parkinson/thérapie , Maladie de Parkinson/physiopathologie , Douleur/étiologie , Douleur/métabolisme , Souris de lignée C57BL , Stimulation cérébrale profonde/méthodes , Modèles animaux de maladie humaine , Lévodopa/pharmacologie , Oxidopamine
4.
Nat Commun ; 15(1): 5206, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38897986

RÉSUMÉ

Disrupted glucose metabolism and protein misfolding are key characteristics of age-related neurodegenerative disorders including Parkinson's disease, however their mechanistic linkage is largely unexplored. The hexosamine biosynthetic pathway utilizes glucose and uridine-5'-triphosphate to generate N-linked glycans required for protein folding in the endoplasmic reticulum. Here we find that Parkinson's patient midbrain cultures accumulate glucose and uridine-5'-triphosphate, while N-glycan synthesis rates are reduced. Impaired glucose flux occurred by selective reduction of the rate-limiting enzyme, GFPT2, through disrupted signaling between the unfolded protein response and the hexosamine pathway. Failure of the unfolded protein response and reduced N-glycosylation caused immature lysosomal hydrolases to misfold and accumulate, while accelerating glucose flux through the hexosamine pathway rescued hydrolase function and reduced pathological α-synuclein. Our data indicate that the hexosamine pathway integrates glucose metabolism with lysosomal activity, and its failure in Parkinson's disease occurs by uncoupling of the unfolded protein response-hexosamine pathway axis. These findings offer new methods to restore proteostasis by hexosamine pathway enhancement.


Sujet(s)
Voies de biosynthèse , Glucose , Hexosamine , Cellules souches pluripotentes induites , Lysosomes , Mésencéphale , Neurones , Maladie de Parkinson , Réponse aux protéines mal repliées , Humains , Hexosamine/biosynthèse , Hexosamine/métabolisme , Lysosomes/métabolisme , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Neurones/métabolisme , Cellules souches pluripotentes induites/métabolisme , Mésencéphale/métabolisme , Glucose/métabolisme , Glycosylation , alpha-Synucléine/métabolisme , Glutamine fructose 6-phosphate transaminase (isomerizing)/métabolisme , Glutamine fructose 6-phosphate transaminase (isomerizing)/génétique
5.
Brain Behav ; 14(6): e3573, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38898625

RÉSUMÉ

INTRODUCTION: Anorexia nervosa (AN) is a debilitating and potentially chronic eating disorder, characterized by low hedonic drive toward food, which has been linked with perturbations in both reward processing and dopaminergic activity. Neuromelanin-sensitive magnetic resonance imaging (MRI) is an emerging method to index midbrain neuromelanin-a by-product of dopaminergic synthesis. The assessment of midbrain neuromelanin, and its association with AN psychopathology and reward-related processes, may provide critical insights into reward circuit function in AN. METHODS: This study will incorporate neuromelanin-sensitive MRI into an existing study of appetitive conditioning in those with AN. Specifically, those with acute and underweight AN (N = 30), those with weight-restored AN (N = 30), and age-matched healthy controls (N = 30) will undergo clinical assessment of current and previous psychopathology, in addition to structural neuromelanin-sensitive MRI, diffusion MRI, and functional MRI (fMRI) during appetitive conditioning. CONCLUSION: This study will be among the first to interrogate midbrain neuromelanin in AN-a disorder characterized by altered dopaminergic activity. Results will help establish whether abnormalities in the midbrain synthesis of dopamine are evident in those with AN and are associated with symptomatic behavior and reduced ability to experience pleasure and reward.


Sujet(s)
Anorexie mentale , Imagerie par résonance magnétique , Mélanines , Mésencéphale , Récompense , Humains , Mélanines/métabolisme , Anorexie mentale/imagerie diagnostique , Anorexie mentale/métabolisme , Anorexie mentale/physiopathologie , Mésencéphale/imagerie diagnostique , Mésencéphale/métabolisme , Imagerie par résonance magnétique/méthodes , Femelle , Adulte , Jeune adulte , Adolescent , Mâle ,
6.
Cell Rep ; 43(5): 114187, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38722743

RÉSUMÉ

The locomotor role of dopaminergic neurons is traditionally attributed to their ascending projections to the basal ganglia, which project to the mesencephalic locomotor region (MLR). In addition, descending dopaminergic projections to the MLR are present from basal vertebrates to mammals. However, the neurons targeted in the MLR and their behavioral role are unknown in mammals. Here, we identify genetically defined MLR cells that express D1 or D2 receptors and control different motor behaviors in mice. In the cuneiform nucleus, D1-expressing neurons promote locomotion, while D2-expressing neurons stop locomotion. In the pedunculopontine nucleus, D1-expressing neurons promote locomotion, while D2-expressing neurons evoke ipsilateral turns. Using RNAscope, we show that MLR dopamine-sensitive neurons comprise a combination of glutamatergic, GABAergic, and cholinergic neurons, suggesting that different neurotransmitter-based cell types work together to control distinct behavioral modules. Altogether, our study uncovers behaviorally relevant cell types in the mammalian MLR based on the expression of dopaminergic receptors.


Sujet(s)
Dopamine , Neurones dopaminergiques , Locomotion , Mésencéphale , Récepteur dopamine D1 , Animaux , Mésencéphale/métabolisme , Souris , Neurones dopaminergiques/métabolisme , Dopamine/métabolisme , Récepteur dopamine D1/métabolisme , Récepteur D2 de la dopamine/métabolisme , Souris de lignée C57BL , Neurones cholinergiques/métabolisme , Neurones cholinergiques/physiologie , Neurones GABAergiques/métabolisme , Mâle
7.
Stem Cell Reports ; 19(6): 830-838, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38759646

RÉSUMÉ

The differentiation of human pluripotent stem cells into ventral mesencephalic dopaminergic (DA) fate is relevant for the treatment of Parkinson's disease. Shortcuts to obtaining DA cells through direct reprogramming often include forced expression of the transcription factor LMX1A. Although reprogramming with LMX1A can generate tyrosine hydroxylase (TH)-positive cells, their regional identity remains elusive. Using an in vitro model of early human neural tube patterning, we report that forced LMX1A expression induced a ventral-to-dorsal fate shift along the entire neuroaxis with the emergence of roof plate fates despite the presence of ventralizing molecules. The LMX1A-expressing progenitors gave rise to grafts containing roof plate-derived choroid plexus cysts as well as ectopically induced TH-positive neurons of a forebrain identity. Early activation of LMX1A prior to floor plate specification was necessary for the dorsalizing effect. Our work suggests using caution in employing LMX1A for the induction of DA fate, as this factor may generate roof plate rather than midbrain fates.


Sujet(s)
Différenciation cellulaire , Neurones dopaminergiques , Cellules souches embryonnaires humaines , Protéines à homéodomaine LIM , Mésencéphale , Facteurs de transcription , Humains , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/cytologie , Protéines à homéodomaine LIM/métabolisme , Protéines à homéodomaine LIM/génétique , Mésencéphale/cytologie , Mésencéphale/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Cellules souches embryonnaires humaines/métabolisme , Cellules souches embryonnaires humaines/cytologie , Plan d'organisation du corps/génétique , Tyrosine 3-monooxygenase/métabolisme , Tyrosine 3-monooxygenase/génétique , Animaux , Régulation de l'expression des gènes au cours du développement
8.
Sci Adv ; 10(22): eadn4203, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38809978

RÉSUMÉ

Learning causal relationships relies on understanding how often one event precedes another. To investigate how dopamine neuron activity and neurotransmitter release change when a retrospective relationship is degraded for a specific pair of events, we used outcome-selective Pavlovian contingency degradation in rats. Conditioned responding was attenuated for the cue-reward contingency that was degraded, as was dopamine neuron activity in the midbrain and dopamine release in the ventral striatum in response to the cue and subsequent reward. Contingency degradation also abolished the trial-by-trial history dependence of the dopamine responses at the time of trial outcome. This profile of changes in cue- and reward-evoked responding is not easily explained by a standard reinforcement learning model. An alternative model based on learning causal relationships was better able to capture dopamine responses during contingency degradation, as well as conditioned behavior following optogenetic manipulations of dopamine during noncontingent rewards. Our results suggest that mesostriatal dopamine encodes the contingencies between meaningful events during learning.


Sujet(s)
Signaux , Dopamine , Neurones dopaminergiques , Récompense , Animaux , Dopamine/métabolisme , Rats , Mâle , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/physiologie , Conditionnement classique , Striatum ventral/métabolisme , Striatum ventral/physiologie , Apprentissage/physiologie , Mésencéphale/métabolisme , Mésencéphale/physiologie ,
9.
Sci Total Environ ; 934: 173119, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38750743

RÉSUMÉ

Paraquat (PQ) is a broad-spectrum herbicide used worldwide and is a hazardous chemical to human health. Cumulative evidence strengthens the association between PQ exposure and the development of Parkinson's disease (PD). However, the underlying mechanism and effective interventions against PQ-induced neurotoxicity remain unclear. In this study, C57BL/6 J mice were treated with PQ (i.p., 10 mg/kg, twice a week) and melatonin (i.g., 20 mg/kg, twice a week) for 8 weeks. Results showed that PQ-induced motor deficits and midbrain dopaminergic neuronal damage in C57BL/6 J mice were protected by melatonin pretreatment. In isolated primary midbrain neurons and SK-N-SH cells, reduction of cell viability, elevation of total ROS levels, axonal mitochondrial transport defects and mitochondrial dysfunction caused by PQ were attenuated by melatonin. After screening of expression of main motors driving axonal mitochondrial transport, data showed that PQ-decreased KIF5A expression in mice midbrain and in SK-N-SH cell was antagonized by melatonin. Using the in vitro KIF5A-overexpression model, it was found that KIF5A overexpression inhibited PQ-caused neurotoxicity and mitochondrial dysfunction in SK-N-SH cells. In addition, application of MTNR1B (MT2) receptor antagonist, 4-P-PDOT, significantly counteracted the protection of melatonin against PQ-induced neurotoxicity. Further, Kif5a-knockdown diminished melatonin-induced alleviation of motor deficits and neuronal damage against PQ in C57BL/6 J mice. The present study establishes a causal link between environmental neurotoxicants exposure and PD etiology and provides effective interventive targets in the pathogenesis of PD.


Sujet(s)
Kinésine , Mélatonine , Mésencéphale , Souris de lignée C57BL , Mitochondries , Paraquat , Paraquat/toxicité , Animaux , Mélatonine/pharmacologie , Souris , Mésencéphale/effets des médicaments et des substances chimiques , Mésencéphale/métabolisme , Kinésine/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Herbicides/toxicité , Neurones/effets des médicaments et des substances chimiques , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Transport axonal/effets des médicaments et des substances chimiques
10.
Alcohol Alcohol ; 59(4)2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38742547

RÉSUMÉ

AIMS: Continued alcohol consumption despite negative consequences is a core symptom of alcohol use disorder. This is modeled in mice by pairing negative stimuli with alcohol, such as adulterating alcohol solution with quinine. Mice consuming alcohol under these conditions are considered to be engaging in aversion-resistant intake. Previously, we have observed sex differences in this behavior, with females more readily expressing aversion-resistant consumption. We also identified three brain regions that exhibited sex differences in neuronal activation during quinine-alcohol drinking: ventromedial prefrontal cortex (vmPFC), posterior insular cortex (PIC), and ventral tegmental area (VTA). Specifically, male mice showed increased activation in vmPFC and PIC, while females exhibited increased activation in VTA. In this study, we aimed to identify what specific type of neurons are activated in these regions during quinine-alcohol drinking. METHOD: We assessed quinine-adulterated alcohol intake using the two-bottle choice procedure. We also utilized RNAscope in situ hybridization in the three brain regions that previously exhibited a sex difference to examine colocalization of Fos, glutamate, GABA, and dopamine. RESULT: Females showed increased aversion-resistant alcohol consumption compared to males. We also found that males had higher colocalization of glutamate and Fos in vmPFC and PIC, while females had greater dopamine and Fos colocalization in the VTA. CONCLUSIONS: Collectively, these experiments suggest that glutamatergic output from the vmPFC and PIC may have a role in suppressing, and dopaminergic activity in the VTA may promote, aversion-resistant alcohol consumption. Future experiments will examine neuronal circuits that contribute to sex differences in aversion resistant consumption.


Sujet(s)
Consommation d'alcool , Neurones , Quinine , Caractères sexuels , Animaux , Quinine/pharmacologie , Femelle , Mâle , Souris , Neurones/effets des médicaments et des substances chimiques , Aire tegmentale ventrale/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Cortex préfrontal/effets des médicaments et des substances chimiques , Mésencéphale/métabolisme , Mésencéphale/effets des médicaments et des substances chimiques , Cortex insulaire/effets des médicaments et des substances chimiques , Cortex cérébral/effets des médicaments et des substances chimiques , Cortex cérébral/métabolisme , Éthanol/pharmacologie , Acide glutamique/métabolisme
11.
Sci Rep ; 14(1): 10983, 2024 05 14.
Article de Anglais | MEDLINE | ID: mdl-38744869

RÉSUMÉ

Parkinson's disease (PD) is a complex neurodegenerative disorder without a cure. The onset of PD symptoms corresponds to 50% loss of midbrain dopaminergic (mDA) neurons, limiting early-stage understanding of PD. To shed light on early PD development, we study time series scRNA-seq datasets of mDA neurons obtained from patient-derived induced pluripotent stem cell differentiation. We develop a new data integration method based on Non-negative Matrix Tri-Factorization that integrates these datasets with molecular interaction networks, producing condition-specific "gene embeddings". By mining these embeddings, we predict 193 PD-related genes that are largely supported (49.7%) in the literature and are specific to the investigated PINK1 mutation. Enrichment analysis in Kyoto Encyclopedia of Genes and Genomes pathways highlights 10 PD-related molecular mechanisms perturbed during early PD development. Finally, investigating the top 20 prioritized genes reveals 12 previously unrecognized genes associated with PD that represent interesting drug targets.


Sujet(s)
Neurones dopaminergiques , Maladie de Parkinson , Maladie de Parkinson/génétique , Maladie de Parkinson/anatomopathologie , Humains , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/anatomopathologie , RNA-Seq/méthodes , Cellules souches pluripotentes induites/métabolisme , Mésencéphale/métabolisme , Mésencéphale/anatomopathologie , Réseaux de régulation génique , Mutation , Différenciation cellulaire/génétique , Multi-omique , Analyse de l'expression du gène de la cellule unique
12.
J Parkinsons Dis ; 14(4): 681-692, 2024.
Article de Anglais | MEDLINE | ID: mdl-38578903

RÉSUMÉ

Background: Alpha-synuclein (aSyn) is a key player in neurodegenerative diseases such as Parkinson's disease (PD), dementia with Lewy bodies, or multiple system atrophy. aSyn is expressed throughout the brain, and can also be detected in various peripheral tissues. In fact, initial symptoms of PD are non-motoric and include autonomic dysfunction, suggesting that the periphery might play an important role in early development of the disease. aSyn is expressed at relatively low levels in non-central tissues, which brings challenges for its detection and quantification in different tissues. Objective: Our goal was to assess the sensitivity of aSyn detection in central and peripheral mouse tissues through capillary electrophoresis (CE) immunoblot, considering the traditional SDS-PAGE immunoblot as the current standard. Methods: Tissues from central and non-central origin from wild type mice were extracted, and included midbrain, inner ear, and esophagus/stomach. aSyn detection was assessed through immunoblotting using Simple Western size-based CE and SDS-PAGE. Results: CE immunoblots show a consistent detection of aSyn in central and peripheral tissues. Through SDS-PAGE, immunoblots revealed a reliable signal corresponding to aSyn, particularly following membrane fixation. Conclusion: Our results suggest a reliable detection of aSyn in central and peripheral tissues using the CE Simple Western immunoblot system. These observations can serve as preliminary datasets when aiming to formally compare CE with SDS-PAGE, as well as for further characterization of aSyn using this technique.


Sujet(s)
Électrophorèse capillaire , alpha-Synucléine , Animaux , alpha-Synucléine/métabolisme , alpha-Synucléine/analyse , Souris , Électrophorèse capillaire/méthodes , Souris de lignée C57BL , Immunotransfert/méthodes , Oesophage/métabolisme , Mésencéphale/métabolisme
13.
eNeuro ; 11(5)2024 May.
Article de Anglais | MEDLINE | ID: mdl-38658137

RÉSUMÉ

The primary motor cortex (M1) integrates sensory and cognitive inputs to generate voluntary movement. Its functional impairments have been implicated in the pathophysiology of motor symptoms in Parkinson's disease (PD). Specifically, dopaminergic degeneration and basal ganglia dysfunction entrain M1 neurons into the abnormally synchronized bursting pattern of activity throughout the cortico-basal ganglia-thalamocortical network. However, how degeneration of the midbrain dopaminergic neurons affects the anatomy, microcircuit connectivity, and function of the M1 network remains poorly understood. The present study examined whether and how the loss of dopamine (DA) affects the morphology, cellular excitability, and synaptic physiology of Layer 5 parvalbumin-expressing (PV+) cells in the M1 of mice of both sexes. Here, we reported that loss of midbrain dopaminergic neurons does not alter the number, morphology, and physiology of Layer 5 PV+ cells in M1. Moreover, we demonstrated that the number of perisomatic PV+ puncta of M1 pyramidal neurons as well as their functional innervation of cortical pyramidal neurons were not altered following the loss of DA. Together, the present study documents an intact GABAergic inhibitory network formed by PV+ cells following the loss of midbrain dopaminergic neurons.


Sujet(s)
Neurones dopaminergiques , Interneurones , Mésencéphale , Cortex moteur , Parvalbumines , Animaux , Femelle , Mâle , Souris , Neurones dopaminergiques/métabolisme , Neurones GABAergiques/métabolisme , Interneurones/métabolisme , Mésencéphale/métabolisme , Souris de lignée C57BL , Souris transgéniques , Cortex moteur/métabolisme , Inhibition nerveuse/physiologie , Parvalbumines/métabolisme
14.
Life Sci ; 345: 122610, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38580194

RÉSUMÉ

Brain Organiods (BOs) are a promising technique for researching disease progression in the human brain. These organoids, which are produced from human induced pluripotent stem cells (HiPSCs), can construct themselves into structured frameworks. In the context of Parkinson's disease (PD), recent advancements have been made in the development of Midbrain organoids (MBOs) models that consider key pathophysiological mechanisms such as alpha-synuclein (α-Syn), Lewy bodies, dopamine loss, and microglia activation. However, there are limitations to the current use of BOs in disease modelling and drug discovery, such as the lack of vascularization, long-term differentiation, and absence of glial cells. To address these limitations, researchers have proposed the use of spinning bioreactors to improve oxygen and nutrient perfusion. Modelling PD utilising modern experimental in vitro models is a valuable tool for studying disease mechanisms and elucidating previously unknown features of PD. In this paper, we exclusively review the unique methods available for cultivating MBOs using a pumping system that mimics the circulatory system. This mechanism may aid in delivering the required amount of oxygen and nutrients to all areas of the organoids, preventing cell death, and allowing for long-term culture and using co-culturing techniques for developing glial cell in BOs. Furthermore, we emphasise some of the significant discoveries about the BOs and the potential challenges of using BOs will be discussed.


Sujet(s)
Cellules souches pluripotentes induites , Maladie de Parkinson , Humains , Maladie de Parkinson/métabolisme , alpha-Synucléine/métabolisme , Mésencéphale/métabolisme , Mésencéphale/anatomopathologie , Organoïdes/métabolisme , Oxygène/métabolisme , Neurones dopaminergiques/métabolisme
15.
Zhen Ci Yan Jiu ; 49(4): 384-390, 2024 Apr 25.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38649206

RÉSUMÉ

OBJECTIVES: To observe the effects on tyrosine hydroxylase (TH), α-synaptic nucleoprotein (α-syn), sirtuin 3 (Sirt3), NOD-like receptor 3 (NLRP3) and gasdermin-D (GSDMD) in the substantia nigra of midbrain after electroacupuncture (EA) at "Fengfu"(GV16), "Taichong" (LR3) and "Zusanli" (ST36) in rats of Parkinson's disease (PD), so as to explore the mechanism of EA in treatment of PD. METHODS: SD rats were randomly divided into control, model and EA groups, with 10 rats in each group. The PD model was established by injecting rotenone into the neck and back, lasting 28 days. In the EA group, EA was applied to GV16, LR3 and ST36, 30 min each time, once daily, consecutively for 28 days. The open-field test was adopted to detect the total distance of autonomic movement of rats, and the pole climbing test was used to detect the body coordination ability of rats. In the substania nigra of midbrain, the positive expression of TH was determined using immunohistochemistry, the mRNA expression levels of α - syn, Sirt3, NLRP3 and GSDMD were detected by quantitative real-time fluorescence PCR, and the protein expression levels of NLRP3, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) and cysteinyl aspartate specific proteinase (Caspase)-1 were detected by Western blot. RESULTS: Compared with the control group, the total distance of autonomous movement was decreased (P<0.01) in the model group, and the score of pole climbing experiment was increased (P<0.01);in the midbrain substantia nigra the positive expression of TH was decreased (P<0.01);the mRNA expression level of Sirt3 was decreased (P<0.01), and those of α-syn, NLRP3 and GSDMD were increased (P<0.01);while the protein expression levels of NLRP3, ASC and Caspase-1 were increased (P<0.01). When compared with the model group, the total distance of autonomous movement in open field experiment was increased (P<0.01) in the EA group and the score of pole climbing experiment was lower (P<0.05);in the midbrain substantia nigra the positive expression of TH was increased (P<0.01);the mRNA expression level of Sirt3 in the midbrain substantia nigra was increased (P<0.01), and those of α-syn, NLRP3 and GSDMD were reduced (P<0.01);while the protein expression levels of NLRP3, ASC and Caspase-1 decreased (P<0.01, P<0.05). CONCLUSIONS: EA at "GV16" "LR3" and "ST36" can repair the neuronal injury, clear the abnormal accumulation of α-syn in the substania nigra of midbrain, and ameliorate mitochondrial damage in PD rats, which may be obtained by regulating Sirt3/NLRP3/GSDMD signaling pathway, so as to delay the occurrence and development of Parkinson's disease.


Sujet(s)
Électroacupuncture , Protéine-3 de la famille des NLR contenant un domaine pyrine , Maladie de Parkinson , Rat Sprague-Dawley , Transduction du signal , Sirtuine-3 , Sirtuines , Substantia nigra , Animaux , Rats , Points d'acupuncture , Mésencéphale/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Maladie de Parkinson/métabolisme , Maladie de Parkinson/thérapie , Maladie de Parkinson/génétique , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Substantia nigra/métabolisme
16.
Cell Death Dis ; 15(4): 246, 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38575601

RÉSUMÉ

Parkinson's disease (PD) is a debilitating neurodegenerative disease characterized by the loss of midbrain dopaminergic neurons (DaNs) and the abnormal accumulation of α-Synuclein (α-Syn) protein. Currently, no treatment can slow nor halt the progression of PD. Multiplications and mutations of the α-Syn gene (SNCA) cause PD-associated syndromes and animal models that overexpress α-Syn replicate several features of PD. Decreasing total α-Syn levels, therefore, is an attractive approach to slow down neurodegeneration in patients with synucleinopathy. We previously performed a genetic screen for modifiers of α-Syn levels and identified CDK14, a kinase of largely unknown function as a regulator of α-Syn. To test the potential therapeutic effects of CDK14 reduction in PD, we ablated Cdk14 in the α-Syn preformed fibrils (PFF)-induced PD mouse model. We found that loss of Cdk14 mitigates the grip strength deficit of PFF-treated mice and ameliorates PFF-induced cortical α-Syn pathology, indicated by reduced numbers of pS129 α-Syn-containing cells. In primary neurons, we found that Cdk14 depletion protects against the propagation of toxic α-Syn species. We further validated these findings on pS129 α-Syn levels in PD patient neurons. Finally, we leveraged the recent discovery of a covalent inhibitor of CDK14 to determine whether this target is pharmacologically tractable in vitro and in vivo. We found that CDK14 inhibition decreases total and pathologically aggregated α-Syn in human neurons, in PFF-challenged rat neurons and in the brains of α-Syn-humanized mice. In summary, we suggest that CDK14 represents a novel therapeutic target for PD-associated synucleinopathy.


Sujet(s)
Maladies neurodégénératives , Maladie de Parkinson , Synucléinopathies , Animaux , Humains , Souris , Rats , alpha-Synucléine/génétique , alpha-Synucléine/métabolisme , Kinases cyclines-dépendantes/génétique , Kinases cyclines-dépendantes/métabolisme , Neurones dopaminergiques/métabolisme , Mésencéphale/métabolisme , Maladies neurodégénératives/métabolisme , Maladie de Parkinson/traitement médicamenteux , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Synucléinopathies/métabolisme , Synucléinopathies/anatomopathologie
17.
Adv Sci (Weinh) ; 11(21): e2400847, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38549185

RÉSUMÉ

Understanding the impact of long-term opioid exposure on the embryonic brain is critical due to the surging number of pregnant mothers with opioid dependency. However, this has been limited by human brain inaccessibility and cross-species differences in animal models. Here, a human midbrain model is established that uses hiPSC-derived midbrain organoids to assess cell-type-specific responses to acute and chronic fentanyl treatment and fentanyl withdrawal. Single-cell mRNA sequencing of 25,510 cells from organoids in different treatment groups reveals that chronic fentanyl treatment arrests neuronal subtype specification during early midbrain development and alters synaptic activity and neuron projection. In contrast, acute fentanyl treatment increases dopamine release but does not significantly alter gene expression related to cell lineage development. These results provide the first examination of the effects of opioid exposure on human midbrain development at the single-cell level.


Sujet(s)
Analgésiques morphiniques , Mésencéphale , Organoïdes , Humains , Mésencéphale/effets des médicaments et des substances chimiques , Mésencéphale/métabolisme , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/métabolisme , Analgésiques morphiniques/pharmacologie , Fentanyl/pharmacologie , Neurogenèse/effets des médicaments et des substances chimiques
18.
Neuroscience ; 546: 1-19, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38522661

RÉSUMÉ

Midbrain dopaminergic (mDA) neurons are significantly impaired in patients inflicted with Parkinson's disease (PD), subsequently affecting a variety of motor functions. There are four pathways through which dopamine elicits its function, namely, nigrostriatal, mesolimbic, mesocortical and tuberoinfundibular dopamine pathways. SHH and Wnt signalling pathways in association with favourable expression of a variety of genes, promotes the development and differentiation of mDA neurons in the brain. However, there is a knowledge gap regarding the complex signalling pathways involved in development of mDA neurons. hiPSC models have been acclaimed to be effective in generating complex disease phenotypes. These models mimic the microenvironment found in vivo thus ensuring maximum reliability. Further, a variety of therapeutic compounds can be screened using hiPSCs since they can be used to generate neurons that could carry an array of mutations associated with both familial and sporadic PD. Thus, culturing hiPSCs to study gene expression and dysregulation of cellular processes associated with PD can be useful in developing targeted therapies that will be a step towards halting disease progression.


Sujet(s)
Neurones dopaminergiques , Cellules souches pluripotentes induites , Mésencéphale , Maladie de Parkinson , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/anatomopathologie , Humains , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Mésencéphale/métabolisme , Mésencéphale/anatomopathologie , Cellules souches pluripotentes induites/métabolisme , Animaux , Différenciation cellulaire/physiologie
19.
Nat Aging ; 4(3): 364-378, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38491288

RÉSUMÉ

Age is the primary risk factor for Parkinson's disease (PD), but how aging changes the expression and regulatory landscape of the brain remains unclear. Here we present a single-nuclei multiomic study profiling shared gene expression and chromatin accessibility of young, aged and PD postmortem midbrain samples. Combined multiomic analysis along a pseudopathogenesis trajectory reveals that all glial cell types are affected by age, but microglia and oligodendrocytes are further altered in PD. We present evidence for a disease-associated oligodendrocyte subtype and identify genes lost over the aging and disease process, including CARNS1, that may predispose healthy cells to develop a disease-associated phenotype. Surprisingly, we found that chromatin accessibility changed little over aging or PD within the same cell types. Peak-gene association patterns, however, are substantially altered during aging and PD, identifying cell-type-specific chromosomal loci that contain PD-associated single-nucleotide polymorphisms. Our study suggests a previously undescribed role for oligodendrocytes in aging and PD.


Sujet(s)
Maladie de Parkinson , Humains , Sujet âgé , Maladie de Parkinson/génétique , Multi-omique , Mésencéphale/métabolisme , Microglie/métabolisme , Noyau du tractus solitaire/métabolisme , Chromatine
20.
Brain Behav Immun ; 117: 356-375, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38320681

RÉSUMÉ

Both exogenous gaseous and liquid forms of formaldehyde (FA) can induce depressive-like behaviors in both animals and humans. Stress and neuronal excitation can elicit brain FA generation. However, whether endogenous FA participates in depression occurrence remains largely unknown. In this study, we report that midbrain FA derived from lipopolysaccharide (LPS) is a direct trigger of depression. Using an acute depressive model in mice, we found that one-week intraperitoneal injection (i.p.) of LPS activated semicarbazide-sensitive amine oxidase (SSAO) leading to FA production from the midbrain vascular endothelium. In both in vitro and in vivo experiments, FA stimulated the production of cytokines such as IL-1ß, IL-6, and TNF-α. Strikingly, one-week microinfusion of FA as well as LPS into the midbrain dorsal raphe nucleus (DRN, a 5-HT-nergic nucleus) induced depressive-like behaviors and concurrent neuroinflammation. Conversely, NaHSO3 (a FA scavenger), improved depressive symptoms associated with a reduction in the levels of midbrain FA and cytokines. Moreover, the chronic depressive model of mice injected with four-week i.p. LPS exhibited a marked elevation in the levels of midbrain LPS accompanied by a substantial increase in the levels of FA and cytokines. Notably, four-week i.p. injection of FA as well as LPS elicited cytokine storm in the midbrain and disrupted the blood-brain barrier (BBB) by activating microglia and reducing the expression of claudin 5 (CLDN5, a protein with tight junctions in the BBB). However, the administration of 30 nm nano-packed coenzyme-Q10 (Q10, an endogenous FA scavenger), phototherapy (PT) utilizing 630-nm red light to degrade FA, and the combination of PT and Q10, reduced FA accumulation and neuroinflammation in the midbrain. Moreover, the combined therapy exhibited superior therapeutic efficacy in attenuating depressive symptoms compared to individual treatments. Thus, LPS-derived FA directly initiates depression onset, thereby suggesting that scavenging FA represents a promising strategy for depression treatment.


Sujet(s)
Dépression , Lipopolysaccharides , Humains , Souris , Animaux , Lipopolysaccharides/pharmacologie , Dépression/traitement médicamenteux , Maladies neuro-inflammatoires , Cytokines/métabolisme , Mésencéphale/métabolisme , Formaldéhyde
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...