Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.778
Filter
1.
Chemosphere ; 364: 143303, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39251157

ABSTRACT

In organisms, long-term nanopolystyrenes (PS-NPs) exposure can cause toxicity, including neurotoxicity. Quercetin, the flavonol with extensive distribution within plants, possesses diverse biological activities. Nevertheless, the possible effect of quercetin to suppress PS-NPs-induced neurotoxicity and its associated mechanism remains unknown. Thus, in the present work, Caenorhabditis elegans was utilized as the model animal to investigate quercetin's pharmacological effect on suppressing PS-NPs-induced neurotoxicity and the underlying mechanism. PS-NPs exposure at 1-100 µg/L remarkably reduced locomotion behavior, while only PS-NPs exposure at 100 µg/L significantly decrease sensory perception behavior. Meanwhile, the increase in the number of worms with dopaminergic neurodegeneration was detected in nematodes exposed to 100 µg/L PS-NPs and the decreased dopamine content was observed within nematodes exposed to 10-100 µg/L PS-NPs, demonstrating the function of dopaminergic neurodegeneration and disruption of dopamine metabolism in inducing PS-NPs toxicity on neuron capacity. After 100 µg/L PS-NPs exposure, the 25-100 µM quercetin treatment effectively increased the locomotion behavior and the sensory perception behavior. Developmentally, quercetin treatment (100 µM) remarkably enhanced fluorescence intensity while decreasing worm number with neurodegeneration within BZ555 transgenic strains exposed to 100 µg/L PS-NPs. Physiologically, quercetin treatment (100 µM) significantly enhanced dopamine content within nematodes exposed to 100 µg/L PS-NPs. Molecularly, quercetin treatment (100 µM) notably decreased the expressions of genes governing neurodegeneration (mec-4, deg-3, unc-68, itr-1, clp-1, and asp-3) while significantly increasing the expression of genes governing dopamine metabolism (cat-2, cat-1, dop-1, dop-2, dop-3). As revealed by molecular docking results, quercetin might bind to excitotoxic-like ion channels receptors (MEC-4 and DEG-3) and dopamine secreted protein (CAT-2). Consequently, findings in this work demonstrated that long-term PS-NPs exposure within the µg/L range (1-100 µg/L) was toxic to neuron capacity, which was associated with the enhancement in dopaminergic neurodegeneration and disruption of dopamine metabolism. Notably, PS-NPs-mediated neurotoxicity to nematodes is probably suppressed through subsequent quercetin treatment.


Subject(s)
Caenorhabditis elegans , Dopamine , Dopaminergic Neurons , Nanoparticles , Polystyrenes , Quercetin , Animals , Caenorhabditis elegans/drug effects , Quercetin/pharmacology , Dopamine/metabolism , Nanoparticles/toxicity , Polystyrenes/toxicity , Dopaminergic Neurons/drug effects , Locomotion/drug effects
2.
Article in English | MEDLINE | ID: mdl-39231689

ABSTRACT

BACKGROUND: Atrazine (ATR), a commonly used herbicide, is linked to dopaminergic neurotoxicity, which may cause symptoms resembling Parkinson's disease (PD). This study aims to reveal the molecular regulatory networks responsible for ATR exposure and its effects on dopaminergic neurotoxicity based on an integration strategy. METHODS: Our approach involved network toxicology, construction of protein-protein interaction (PPI) networks, gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, as well as molecular docking techniques. Subsequently, we validated the predicted results in PC12 cells in vitro. RESULTS: An integrated analysis strategy indicating that 5 hub targets, including mitogen-activated protein kinase 3 (Mapk3), catalase (Cat), heme oxygenase 1 (Hmox1), tumor protein p53 (Tp53), and prostaglandin-endoperoxide synthase 2 (Ptgs2), may play a crucial role in ATR-induced dopaminergic injury. Molecular docking indicated that the 5 hub targets exhibited certain binding activity with ATR. Cell counting kit-8 (CCK8) results illustrated a dose-response relationship in PC12 cells. Real-time quantitative polymerase chain reaction (RT-qPCR) displayed notable changes in the expression of hub targets mRNA levels, with the exception of Mapk3. Western blotting results suggested that ATR treatment in PC12 cells resulted in an upregulation of the Cat, Hmox1, and p-Mapk3 protein expression levels while causing a downregulation in Tp53, Ptgs2, and Mapk3. CONCLUSION: Our findings indicated that 5 hub targets identified could play a vital role in ATR-induced dopaminergic neurotoxicity in PC12 cells. These results provide preliminary support for further investigation into the molecular mechanism of ATR-induced toxicity.


Subject(s)
Atrazine , Dopaminergic Neurons , Herbicides , Molecular Docking Simulation , Atrazine/toxicity , Animals , PC12 Cells , Rats , Herbicides/toxicity , Dopaminergic Neurons/drug effects , Protein Interaction Maps , Dopamine/metabolism
3.
J Neural Eng ; 21(5)2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39231475

ABSTRACT

Objective.Cryogel microcarriers made of poly(ethylene glycol) diacrylate and 3-sulfopropyl acrylate have the potential to act as delivery vehicles for long-term retention of neurotrophic factors (NTFs) in the brain. In addition, they can potentially enhance stem cell-derived dopaminergic (DAergic) cell replacement strategies for Parkinson's disease (PD), by addressing the limitations of variable survival and poor differentiation of the transplanted precursors due to neurotrophic deprivation post-transplantation in the brain. In this context, to develop a proof-of-concept, the aim of this study was to determine the efficacy of glial cell line-derived NTF (GDNF)-loaded cryogel microcarriers by assessing their impact on the survival of, and reinnervation by, primary DAergic grafts after intra-striatal delivery in Parkinsonian rat brains.Approach.Rat embryonic day 14 ventral midbrain cells were transplanted into the 6-hydroxydopamine-lesioned striatum either alone, or with GDNF, or with unloaded cryogel microcarriers, or with GDNF-loaded cryogel microcarriers.Post-mortem, GDNF and tyrosine hydroxylase immunostaining were used to identify retention of the delivered GDNF within the implanted cryogel microcarriers, and to identify the transplanted DAergic neuronal cell bodies and fibres in the brains, respectively.Main results.We found an intact presence of GDNF-stained cryogel microcarriers in graft sites, indicating their ability for long-term retention of the delivered GDNF up to 4 weeks in the brain. This resulted in an enhanced survival (1.9-fold) of, and striatal reinnervation (density & volume) by, the grafted DAergic neurons, in addition to an enhanced sprouting of fibres within graft sites.Significance.This data provides an important proof-of-principle for the beneficial effects of neurotrophin-loaded cryogel microcarriers on engraftment of cells in the context of cell replacement therapy in PD. For clinical translation, further studies will be needed to assess the impact of cryogel microcarriers on the survival and differentiation of stem cell-derived DAergic precursors in Parkinsonian rat brains.


Subject(s)
Cryogels , Dopaminergic Neurons , Glial Cell Line-Derived Neurotrophic Factor , Animals , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Rats , Cryogels/administration & dosage , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/transplantation , Parkinson Disease/therapy , Rats, Sprague-Dawley , Disease Models, Animal , Cells, Cultured , Male
4.
J Mol Neurosci ; 74(4): 88, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39297981

ABSTRACT

Parkinson's disease (PD) is a common motor neurodegenerative disease that still lacks effective therapeutic options. Previous studies have reported that lactoferrin exhibited neuroprotective effects in cellular and animal models of PD, typically induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or 1-methyl-4-phenylpyridinium (MPP+) synthetic toxin. However, the neuroprotective capacity of lactoferrin in the rotenone-induced cellular model of PD remains relatively less established. Unlike MPTP/MPP+, rotenone is a naturally occurring environmental toxin known to induce chronic toxicity and increase the risk of PD in humans. In this study, we constructed a cellular model of PD by differentiating SH-SY5Y neuroblastoma cells with retinoic acid into mature dopaminergic neurons with increased ß-tubulin III and tyrosine hydroxylase expression, followed by 24 h of rotenone exposure. Using this cellular model of PD, we showed that lactoferrin (1-10 µg/ml) pre-treatment for 48 h decreased loss of cell viability, mitochondrial membrane potential impairment, reactive oxygen species generation and pro-apoptotic activities (pan-caspase activation and nuclear condensation) in cells exposed to rotenone (1 and 5 µM) using biochemical assays, Hoechst 33342 staining and immunocytochemical techniques. We further demonstrated that 48 h of lactoferrin (10 µg/ml) pre-treatment decreased Bax:Bcl2 ratio and p42/44 mitogen-activated protein kinase expression but increased pAkt expression in 5 µM rotenone-exposed cells. Our study demonstrates that lactoferrin neuroprotective capacity is present in the rotenone-induced cellular model of PD, further supporting lactoferrin as a potential PD therapeutic that warrants further studies.


Subject(s)
Apoptosis , Dopaminergic Neurons , Lactoferrin , Neuroprotective Agents , Rotenone , Humans , Rotenone/toxicity , Lactoferrin/pharmacology , Cell Line, Tumor , Neuroprotective Agents/pharmacology , Apoptosis/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Reactive Oxygen Species/metabolism
5.
Cell Death Dis ; 15(8): 625, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39223107

ABSTRACT

Parkinson's Disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the Substantia nigra pars compacta (SNpc). Apoptosis is thought to play a critical role in the progression of PD, and thus understanding the effects of antiapoptotic strategies is crucial for developing potential therapies. In this study, we developed a unique genetic model to selectively delete Casp3, the gene encoding the apoptotic protein caspase-3, in dopaminergic neurons (TH-C3KO) and investigated its effects in response to a subacute regime of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration, which is known to trigger apoptotic loss of SNpc dopaminergic neurons. We found that Casp3 deletion did not protect the dopaminergic system in the long term. Instead, we observed a switch in the cell death pathway from apoptosis in wild-type mice to necrosis in TH-C3KO mice. Notably, we did not find any evidence of necroptosis in our model or in in vitro experiments using primary dopaminergic cultures exposed to 1-methyl-4-phenylpyridinium in the presence of pan-caspase/caspase-8 inhibitors. Furthermore, we detected an exacerbated microglial response in the ventral mesencephalon of TH-C3KO mice in response to MPTP, which mimicked the microglia neurodegenerative phenotype (MGnD). Under these conditions, it was evident the presence of numerous microglial phagocytic cups wrapping around apparently viable dopaminergic cell bodies that were inherently associated with galectin-3 expression. We provide evidence that microglia exhibit phagocytic activity towards both dead and stressed viable dopaminergic neurons through a galectin-3-dependent mechanism. Overall, our findings suggest that inhibiting apoptosis is not a beneficial strategy for treating PD. Instead, targeting galectin-3 and modulating microglial response may be more promising approaches for slowing PD progression.


Subject(s)
Apoptosis , Caspase 3 , Dopaminergic Neurons , Galectin 3 , Microglia , Necrosis , Phagocytosis , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Microglia/metabolism , Microglia/pathology , Microglia/drug effects , Apoptosis/drug effects , Galectin 3/metabolism , Galectin 3/genetics , Caspase 3/metabolism , Mice , Phagocytosis/drug effects , Mice, Knockout , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Mice, Inbred C57BL , Male
6.
Transl Psychiatry ; 14(1): 363, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39242529

ABSTRACT

Reward processing dysfunctions e.g., anhedonia, apathy, are common in stress-related neuropsychiatric disorders including depression and schizophrenia, and there are currently no established therapies. One potential therapeutic approach is restoration of reward anticipation during appetitive behavior, deficits in which co-occur with attenuated nucleus accumbens (NAc) activity, possibly due to NAc inhibition of mesolimbic dopamine (DA) signaling. Targeting NAc regulation of ventral tegmental area (VTA) DA neuron responsiveness to reward cues could involve either the direct or indirect-via ventral pallidium (VP)-pathways. One candidate is the orphan G protein-coupled receptor GPR52, expressed by DA receptor 2 NAc neurons that project to VP. In mouse brain-slice preparations, GPR52 inverse agonist (GPR52-IA) attenuated evoked inhibitory postsynaptic currents at NAc-VP neurons, which could disinhibit VTA DA neurons. A mouse model in which chronic social stress leads to reduced reward learning and effortful motivation was applied to investigate GPR52-IA behavioral effects. Control and chronically stressed mice underwent a discriminative learning test of tone-appetitive behavior-sucrose reinforcement: stress reduced appetitive responding and discriminative learning, and these anticipatory behaviors were dose-dependently reinstated by GPR52-IA. The same mice then underwent an effortful motivation test of operant behavior-tone-sucrose reinforcement: stress reduced effortful motivation and GPR52-IA dose-dependently restored it. In a new cohort, GRABDA-sensor fibre photometry was used to measure NAc DA activity during the motivation test: in stressed mice, reduced motivation co-occurred with attenuated NAc DA activity specifically to the tone that signaled reinforcement of effortful behavior, and GPR52-IA ameliorated both deficits. These findings: (1) Demonstrate preclinical efficacy of GPR52 inverse agonism for stress-related deficits in reward anticipation during appetitive behavior. (2) Suggest that GPR52-dependent disinhibition of the NAc-VP-VTA-NAc circuit, leading to increased phasic NAc DA signaling of earned incentive stimuli, could account for these clinically relevant effects.


Subject(s)
Dopamine , Dopaminergic Neurons , Motivation , Nucleus Accumbens , Reward , Stress, Psychological , Animals , Male , Mice , Behavior, Animal/drug effects , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Mice, Inbred C57BL , Motivation/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Stress, Psychological/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
7.
Adv Sci (Weinh) ; 11(36): e2310244, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39099429

ABSTRACT

The most pronounced neuropathological feature of Parkinson's disease (PD) is the loss of dopamine (DA) neurons in the substantia nigra compacta (SNc), which depletes striatal DA. Hypothalamic oxytocin is found to be reduced in PD patients and closely interacts with the DA system, but the role of oxytocin in PD remains unclear. Here, the disturbances of endogenous oxytocin level and the substantia nigra (SN) oxytocin receptor expression in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model is observed, correlated with the striatal tyrosine hydroxylase (TH) expression reduction. Killing/silencing hypothalamic oxytocin neurons aggravates the vulnerability of nigrostriatal DA signal to MPTP, whereas elevating oxytocin level by intranasal delivery or microinjecting into the SN promotes the resistance. In addition, knocking out SN oxytocin receptors induces the time-dependent reductions of SNc DA neurons, striatal TH expression, and striatal DA level by increasing neuronal excitotoxicity. These results further uncover that oxytocin dampens the excitatory synaptic inputs onto DA neurons via activating oxytocin receptor-expressed SN GABA neurons, which target GABA(B) receptors expressed in SNc DA neuron-projecting glutamatergic axons, to reduce excitotoxicity. Thus, besides the well-known prosocial effect, oxytocin acts as a key endogenous factor in protecting the nigrostriatal DA system.


Subject(s)
Corpus Striatum , Disease Models, Animal , Dopamine , Mice, Inbred C57BL , Oxytocin , Substantia Nigra , Animals , Oxytocin/metabolism , Oxytocin/pharmacology , Mice , Substantia Nigra/metabolism , Substantia Nigra/drug effects , Male , Dopamine/metabolism , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Parkinson Disease/metabolism , Parkinson Disease/genetics , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Receptors, Oxytocin/metabolism , Receptors, Oxytocin/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
8.
J Neurol Sci ; 465: 123188, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39178824

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder resulting from the loss of dopamine-producing neurons in the brain, causing motor symptoms like tremors and stiffness. Although current treatments like medication and deep brain stimulation can alleviate symptoms, they don't address the root cause of neuron loss. Therefore, cell replacement therapy emerges as a promising treatment strategy. However, the generation of engraftable dopaminergic (DA) cells in clinically relevant quantities is still a challenge. Recent advances in cell reprogramming technologies open up vast possibilities to produce patient-specific cells of a desired type in therapeutic quantities. The main cell reprogramming strategies involve the enforced expression of individual or sets of genes through viral transduction or transfection, or through small molecules, known as the chemical approach, which is a much easier and safer method. In our previous studies, using a small molecule approach (combinations of epigenetic modifiers and SMAD inhibitors such asDorsomorphin and SB431542), we have been able to generate DA progenitors from human mesenchymal stem cells (hMSCs). The aim of this study was to further improve the method for the generation of DA progenitors and to test their therapeutic effect in an animal model of Parkinson's. The results showed that the addition of an autophagy enhancer (AE) to our DA cell induction protocol further increased the yield of DA progenitor cells. The results also showed that DA progenitors transplanted into the mouse model of PD survived, integrated, and improved PD motor symptoms. These data suggest that chemically-produced DA cells can be very promising and safe cellular therapeutics for PD.


Subject(s)
Disease Models, Animal , Dopaminergic Neurons , Oxidopamine , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Mice , Oxidopamine/toxicity , Humans , Recovery of Function/drug effects , Recovery of Function/physiology , Parkinsonian Disorders/therapy , Mice, Inbred C57BL , Neural Stem Cells/transplantation , Neural Stem Cells/drug effects , Parkinson Disease/therapy , Parkinson Disease/metabolism , Male , Mesenchymal Stem Cells , Cells, Cultured
9.
J Neurosci ; 44(39)2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39187377

ABSTRACT

Dopamine (DA) neurons in the ventral tegmental area (VTA) respond to motivationally relevant cues, and circuit-specific signaling drives different aspects of motivated behavior. Orexin (ox; also known as hypocretin) and dynorphin (dyn) are coexpressed lateral hypothalamic (LH) neuropeptides that project to the VTA. These peptides have opposing effects on the firing activity of VTADA neurons via orexin 1 (Ox1R) or kappa opioid (KOR) receptors. Given that Ox1R activation increases VTADA firing, and KOR decreases firing, it is unclear how the coreleased peptides contribute to the net activity of DA neurons. We tested if optical stimulation of LHox/dyn neuromodulates VTADA neuronal activity via peptide release and if the effects of optically driven LHox/dyn release segregate based on VTADA projection targets including the basolateral amygdala (BLA) or the lateral or medial shell of the nucleus accumbens (lAcbSh, mAchSh). Using a combination of circuit tracing, optogenetics, and patch-clamp electrophysiology in male and female orexincre mice, we showed a diverse response of LHox/dyn optical stimulation on VTADA neuronal firing, which is not mediated by fast transmitter release and is blocked by antagonists to KOR and Ox1R signaling. Additionally, where optical stimulation of LHox/dyn inputs in the VTA inhibited firing of the majority of BLA-projecting VTADA neurons, optical stimulation of LHox/dyn inputs in the VTA bidirectionally affects firing of either lAcbSh- or mAchSh-projecting VTADA neurons. These findings indicate that LHox/dyn corelease may influence the output of the VTA by balancing ensembles of neurons within each population which contribute to different aspects of reward seeking.


Subject(s)
Dopaminergic Neurons , Dynorphins , Orexins , Ventral Tegmental Area , Animals , Orexins/metabolism , Orexins/pharmacology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology , Dynorphins/metabolism , Dynorphins/pharmacology , Mice , Dopaminergic Neurons/physiology , Dopaminergic Neurons/drug effects , Male , Mice, Inbred C57BL , Female , Neural Pathways/physiology , Neural Pathways/drug effects , Hypothalamic Area, Lateral/physiology , Hypothalamic Area, Lateral/drug effects , Mice, Transgenic , Optogenetics , Orexin Receptors/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology
10.
Front Immunol ; 15: 1410784, 2024.
Article in English | MEDLINE | ID: mdl-39156892

ABSTRACT

Background: PaBing-II Formula (PB-II) is a traditional Chinese medicine for treating Parkinson's disease (PD). However, owing to the complexity of PB-II and the difficulty in obtaining human dopaminergic neurons (DAn), the mechanism of action of PB-II in PD treatment remains unclear. The aim of this study was to investigate the mechanisms underlying the therapeutic benefits of PB-II in patients with PD. Methods: hiPSCs derived DAn were treated with H2O2 to construct the DAn oxidative damage model. SwissTargetPrediction was employed to predict the potential targets of the main compounds in serum after PB-II treatment. Metascape was used to analyze the pathways. Sprague-Dawley rats were used to construct the 6-hydroxydopamine (6-OHDA)-induced PD model, and the duration of administration was four weeks. RNA sequencing was used for Transcriptome analysis to find the signal pathways related to neuronal damage. The associated inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA). We identified PB-II as an Nrf2 activator using antioxidant-responsive element luciferase assay in MDA-MB-231 cells. Results: In vitro experiments showed that the treatment of PB-II-treated serum increased the percentage of TH+ cells, decreased inflammation and the apoptosis, reduced cellular reactive oxygen species, and upregulated the expression of Nrf2 and its downstream genes. Pathway analysis of the RNA-seq data of samples before and after the treatment with PB-II-treated serum identified neuron-associated pathways. In vivo experiments demonstrated that PB-II treatment of PD rat model could activate the Nrf2 signaling pathway, protect the midbrain DAn, and improve the symptoms in PD rats. Conclusion: PB-II significantly protects DAn from inflammation and oxidative stress via Nrf2 pathway activation. These findings elucidate the roles of PB-II in PD treatment and demonstrate the application of hiPSC-derived DAn in research of Chinese medicine.


Subject(s)
Dopaminergic Neurons , Drugs, Chinese Herbal , Induced Pluripotent Stem Cells , NF-E2-Related Factor 2 , Oxidative Stress , Rats, Sprague-Dawley , Humans , Oxidative Stress/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Animals , Drugs, Chinese Herbal/pharmacology , Rats , Induced Pluripotent Stem Cells/metabolism , NF-E2-Related Factor 2/metabolism , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Male , Signal Transduction/drug effects , Disease Models, Animal , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidopamine , Medicine, Chinese Traditional
11.
Dev Psychobiol ; 66(6): e22536, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39164823

ABSTRACT

The synthetic progestin, 17-α-hydroxyprogesterone caproate (17-OHPC), is administered to pregnant individuals at risk for recurrent preterm birth during a critical period of fetal mesocorticolimbic serotonergic and dopaminergic pathway development. These pathways play an important role in regulating cognitive behaviors later in life. Despite this, there has been very little research regarding the potential long-term effects of 17-OHPC on the behavioral and neural development of exposed children. In rodents, developmental exposure to 17-OHPC disrupts serotonergic and dopaminergic innervation of the medial prefrontal cortex and impairs decision-making in complex cognitive tasks in adulthood. The present study tested the hypothesis that developmental exposure to 17-OHPC similarly disrupts the development of serotonergic and dopaminergic pathways within limbic targets and subsequent mood-related behaviors. Developmental 17-OHPC exposure significantly increased the density of serotonin transporter-IR fibers in CA1, CA2/3, and the suprapyramidal blade of dentate gyrus in hippocampus and significantly reduced the density of TH-IR fibers within the nucleus accumbens shell in males but had no effect in females during adolescence. Irregular microglia activational phenotype and number were also observed in the hippocampus of 17-OHPC-exposed males. Developmental 17-OHPC reduced the latency to immobility in males in the forced swim test but did not affect sucrose consumption in a sucrose preference test. These findings suggest that 17-OHPC exerts sex-specific effects on the development of mesocorticolimbic pathways and mood-related behavior in adolescence and highlight the need to investigate effects in adolescent children.


Subject(s)
Behavior, Animal , Animals , Male , Female , Rats , Behavior, Animal/drug effects , Behavior, Animal/physiology , Pregnancy , Affect/drug effects , Affect/physiology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Dopamine/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Serotonin Plasma Membrane Transport Proteins/metabolism , Rats, Sprague-Dawley , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Serotonin/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology
12.
Int J Nanomedicine ; 19: 8463-8483, 2024.
Article in English | MEDLINE | ID: mdl-39185346

ABSTRACT

Introduction: Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra. The precise molecular mechanisms underlying neuronal loss in PD remain unknown, and there are currently no effective treatments for PD-associated neurodegeneration. Echinacoside (ECH) is known for its neuroprotective effects, which include scavenging cellular reactive oxygen species and promoting mitochondrial fusion. However, the blood-brain barrier (BBB) limits the bioavailability of ECH in the brain, posing a significant challenge to its use in PD treatment. Methods: We synthesized and characterized PEGylated ECH liposomes (ECH@Lip) and peptide angiopep-2 (ANG) modified liposomes (ECH@ANG-Lip). The density of ANG in ANG-Lip was optimized using bEnd.3 cells. The brain-targeting ability of the liposomes was assessed in vitro using a transwell BBB model and in vivo using an imaging system and LC-MS. We evaluated the enhanced neuroprotective properties of this formulation in a the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model. Results: The ECH@ANG-Lip demonstrated significantly higher whole-brain uptake compared to ECH@Lip and free ECH. Furthermore, ECH@ANG-Lip was more effective in mitigating MPTP-induced behavioral impairment, oxidative stress, dopamine depletion, and dopaminergic neuron death than both ECH@Lip and free ECH. Conclusion: The formulation used in our study significantly enhanced the neuroprotective efficacy of ECH in the MPTP-induced PD model. Thus, ECH@ANG-Lip shows considerable potential for improving the bioavailability of ECH and providing neuroprotective effects in the brain.


Subject(s)
Blood-Brain Barrier , Disease Models, Animal , Glycosides , Liposomes , Mice, Inbred C57BL , Neuroprotective Agents , Animals , Liposomes/chemistry , Liposomes/pharmacokinetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/pharmacokinetics , Mice , Male , Glycosides/chemistry , Glycosides/pharmacology , Glycosides/pharmacokinetics , Brain/drug effects , Brain/metabolism , Parkinson Disease/drug therapy , Cell Line , Dopaminergic Neurons/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics
13.
Sci Rep ; 14(1): 20123, 2024 08 29.
Article in English | MEDLINE | ID: mdl-39209951

ABSTRACT

Modulation of the Nrf2 pathway, a master regulator of the antioxidant response and cellular metabolism, has been suggested as a promising therapeutic strategy in tauopathies, a heterogeneous group of neurodegenerative disorders characterized by intracellular proteinaceous inclusions of abnormally phosphorylated tau. Here, we explored the neuroprotective potential of different Nrf2-pathway activators in human immortalized dopaminergic neurons against annonacin-induced toxicity, a mitochondrial inhibitor associated with a PSP-like syndrome and capable of mimicking tauopathy-like features. Interestingly, we observed heterogenous and compound-dependent neuroprotective effects among the different Nrf2-pathway activators. With the exception of Fyn inhibitors, all the selected Nrf2-pathway activators improved cell viability and the oxidative status, and reduced the annonacin-induced tau hyperphosphorylation and neurite degeneration, particularly the p62-activators. However, improvement of the impaired mitochondrial function was only observed by the Bach-1 inhibitor. Surprisingly, we found evidence that ezetimibe, an approved drug for hypercholesterolemia, prevents the transcriptional upregulation of 4R-tau triggered by annonacin insult. Overall, our results suggest that the neuroprotective effects of the Nrf2-pathway activators against annonacin toxicity may rely on the specific mechanism of action, intrinsic to each compound, and possibly on the concomitant modulation of additional signaling pathways. Further research will be needed to fully understand how synergistic modulation of metabolic adaptation and cell survival can be exploit to develop new therapeutical strategies for tauopathies and eventually other neurodegenerative diseases.


Subject(s)
Cell Survival , NF-E2-Related Factor 2 , Neuroprotective Agents , Signal Transduction , NF-E2-Related Factor 2/metabolism , Humans , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Cell Survival/drug effects , Oxidative Stress/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , tau Proteins/metabolism , Phosphorylation/drug effects , Cell Line , Benzofurans/pharmacology , Furans , Lactones
14.
CNS Neurosci Ther ; 30(8): e14883, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39097919

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a chronic neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra, which promotes a sustained inflammatory environment in the central nervous system. Regulatory T cells (Tregs) play an important role in the control of inflammation and might play a neuroprotective role. Indeed, a decrease in Treg number and function has been reported in PD. In this context, pramipexole, a dopaminergic receptor agonist used to treat PD symptoms, has been shown to increase peripheral levels of Treg cells and improve their suppressive function. The aim of this work was to determine the effect of pramipexole on immunoregulatory Treg cells and its possible neuroprotective effect on human dopaminergic neurons differentiated from human embryonic stem cells. METHODS: Treg cells were sorted from white blood cells of healthy human donors. Assays were performed with CD3/CD28-activated and non-activated Treg cells treated with pramipexole at concentrations of 2 or 200 ng/mL. These regulatory cells were co-cultured with in vitro-differentiated human dopaminergic neurons in a cytotoxicity assay with 6-hydroxydopamine (6-OHDA). The role of interleukin-10 (IL-10) was investigated by co-culturing activated IL-10-producing Treg cells with neurons. To further investigate the effect of treatment on Tregs, gene expression in pramipexole-treated, CD3/CD28-activated Treg cells was determined by Fluidigm analysis. RESULTS: Pramipexole-treated CD3/CD28-activated Treg cells showed significant protective effects on dopaminergic neurons when challenged with 6-OHDA. Pramipexole-treated activated Treg cells showed neuroprotective capacity through mechanisms involving IL-10 release and the activation of genes associated with regulation and neuroprotection. CONCLUSION: Anti-CD3/CD28-activated Treg cells protect dopaminergic neurons against 6-OHDA-induced damage. In addition, activated, IL-10-producing, pramipexole-treated Tregs also induced a neuroprotective effect, and the supernatants of these co-cultures promoted axonal growth. Pramipexole-treated, activated Tregs altered their gene expression in a concentration-dependent manner, and enhanced TGFß-related dopamine receptor regulation and immune-related pathways. These findings open new perspectives for the development of immunomodulatory therapies for the treatment of PD.


Subject(s)
Benzothiazoles , Dopaminergic Neurons , Oxidopamine , Pramipexole , T-Lymphocytes, Regulatory , Humans , Pramipexole/pharmacology , T-Lymphocytes, Regulatory/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Oxidopamine/toxicity , Benzothiazoles/pharmacology , Coculture Techniques , Interleukin-10/metabolism , Cells, Cultured , Neuroprotective Agents/pharmacology , Dopamine Agonists/pharmacology
15.
Neuromolecular Med ; 26(1): 32, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090268

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disorder marked by the death of dopaminergic neurons in the substantia nigra region of the brain. Aggregation of alpha-synuclein (α-synuclein) is a contributing factor to Parkinson's disease pathogenesis. The objective of this study is to investigate the neuroprotective effects of gut microbes on α-synuclein aggregation using both in silico and in vivo approaches. We focussed on the interaction between α-synuclein and metabolites released by gut bacteria that protect from PD. We employed three probiotic microbe strains against α-synuclein protein: Lactobacillus casei, Escherichia coli, and Bacillus subtilis, with their chosen PDB IDs being Dihydrofolate reductase (3DFR), methionine synthetase (6BM5), and tryptophanyl-tRNA synthetase (3PRH), respectively. Using HEX Dock 6.0 software, we examined the interactions between these proteins. Among the various metabolites, methionine synthetase produced by E. coli showed potential interactions with α-synuclein. To further evaluate the neuroprotective benefits of E. coli, an in vivo investigation was performed using a rotenone-induced Parkinsonian mouse model. The motor function of the animals was assessed through behavioural tests, and oxidative stress and neurotransmitter levels were also examined. The results demonstrated that, compared to the rotenone-induced PD mouse model, the rate of neurodegeneration was considerably reduced in mice treated with E. coli. Additionally, histopathological studies provided evidence of the neuroprotective effects of E. coli. In conclusion, this study lays the groundwork for future research, suggesting that gut bacteria may serve as potential therapeutic agents in the development of medications to treat Parkinson's disease. fig. 1.


Subject(s)
Bacillus subtilis , Escherichia coli , Gastrointestinal Microbiome , Molecular Docking Simulation , Oxidative Stress , Probiotics , Rotenone , alpha-Synuclein , Animals , Mice , Gastrointestinal Microbiome/physiology , Probiotics/therapeutic use , Probiotics/pharmacology , alpha-Synuclein/metabolism , Oxidative Stress/drug effects , Rotenone/toxicity , Lacticaseibacillus casei/physiology , Methionine-tRNA Ligase , Tryptophan-tRNA Ligase/physiology , Male , Tetrahydrofolate Dehydrogenase/metabolism , Computer Simulation , Parkinsonian Disorders/microbiology , Humans , Neuroprotective Agents/therapeutic use , Mice, Inbred C57BL , Disease Models, Animal , Parkinson Disease, Secondary/chemically induced , Dopaminergic Neurons/drug effects , Parkinson Disease/microbiology
16.
Mol Biol Rep ; 51(1): 908, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39141244

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by oxidative stress and neuroinflammation. Sofalcone (SFC), a chalcone derivative known for its antioxidative and anti-inflammatory properties, is widely used clinically as a gastric mucosa protective agent. However, its therapeutic potential in PD remains to be fully explored. In this study, we investigated the neuroprotective effects of SFC in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. METHODS AND RESULTS: We found that SFC ameliorated MPTP-induced motor impairments in mice, as assessed by the rotarod and wire tests. Moreover, SFC administration prevented the loss of dopaminergic neurons and striatal degeneration induced by MPTP. Subsequent investigations revealed that SFC reversed MPTP-induced downregulation of NRF2, reduced elevated levels of reactive oxygen species (ROS) and malondialdehyde (MDA), and increased total antioxidant capacity (TAOC). Furthermore, SFC suppressed MPTP-induced activation of microglia and astrocytes, downregulated the pro-inflammatory cytokine TNF-α, and upregulated the anti-inflammatory cytokine IL-4. Additionally, SFC ameliorated the MPTP-induced downregulation of phosphorylation of Akt at Ser473. CONCLUSIONS: This study provides evidence for the neuroprotective effects of SFC, highlighting its antioxidative and anti-inflammatory properties and its role in Akt activation in the PD model. These findings underscore SFC's potential as a promising therapeutic candidate for PD, warranting further clinical investigation.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Chalcones , Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Animals , Oxidative Stress/drug effects , Mice , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Chalcones/pharmacology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Antioxidants/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Microglia/drug effects , Microglia/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , NF-E2-Related Factor 2/metabolism , Anti-Inflammatory Agents/pharmacology
17.
Food Funct ; 15(18): 9390-9408, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39189380

ABSTRACT

Parkinson's disease (PD) is a chronic progressive neurodegenerative disease that often occurs in older people. Neuroinflammation and oxidative stress are important factors in the development of PD. Gastrointestinal dysfunction is the most common non-motor symptom, and inflammation of the gut, which activates the gut-brain axis, maybe a pathogenic factor. Previous studies have attributed anti-inflammatory and antioxidant effects to Allantoin, but its function and mechanism of action in PD are unclear. This study aimed to investigate the effect and mechanism of Allantoin on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD in mice. Our results showed that Allantoin administration ameliorated motor dysfunction and neuronal damage in mice injected with MPTP by inhibiting neuroinflammation and oxidative damage. Mechanistic studies showed that Allantoin suppresses inflammatory responses by inhibiting the overactivation of the NF-κB and MAPK signaling pathways, as well as oxidative stress by regulating the AKT/Nrf2/HO-1 signaling pathway. Notably, Allantoin also restored intestinal barrier function by modulating the gut microbiota and improving antioxidant and anti-inflammatory capacities to alleviate MPTP-induced motor deficits. In conclusion, the present study shows that the administration of Allantoin attenuated neurodegeneration in mice injected with MPTP by inhibiting neuroinflammation and oxidative stress and modulating the composition of the gut microbiome.


Subject(s)
Allantoin , Dopaminergic Neurons , Gastrointestinal Microbiome , Mice, Inbred C57BL , Oxidative Stress , Animals , Gastrointestinal Microbiome/drug effects , Mice , Oxidative Stress/drug effects , Male , Dopaminergic Neurons/drug effects , Allantoin/pharmacology , Inflammation/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Disease Models, Animal , Antioxidants/pharmacology
18.
Toxicol Sci ; 201(2): 263-281, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38995845

ABSTRACT

Epidemiological studies show that exposure to the organochlorine pesticide dieldrin is associated with an increased risk of Parkinson's disease (PD). Animal studies support a link between developmental dieldrin exposure and increased neuronal susceptibility in the α-synuclein preformed fibril and MPTP models in adult male C57BL/6 mice. In a previous study, we showed that developmental dieldrin exposure was associated with sex-specific changes in DNA modifications within genes related to dopaminergic neuron development and maintenance at 12 wk of age. Here, we used capture hybridization-sequencing with custom baits to interrogate DNA modifications across the entire genetic loci of the previously identified genes at multiple time points-birth, 6, 12, and 36 wk old. We identified largely sex-specific dieldrin-induced changes in DNA modifications at each time point that annotated to pathways important for neurodevelopment, potentially related to critical steps in early neurodevelopment, dopaminergic neuron differentiation, synaptogenesis, synaptic plasticity, and glial-neuron interactions. Despite large numbers of age-specific DNA modifications, longitudinal analysis identified a small number of differential modification of cytosines with dieldrin-induced deflection of epigenetic aging. The sex-specificity of these results adds to evidence that sex-specific responses to PD-related exposures may underly sex-specific differences in disease. Overall, these data support the idea that developmental dieldrin exposure leads to changes in epigenetic patterns that persist after the exposure period and disrupt critical neurodevelopmental pathways, thereby impacting risk of late-life diseases, including PD.


Subject(s)
Dieldrin , Mesencephalon , Mice, Inbred C57BL , Prenatal Exposure Delayed Effects , Animals , Dieldrin/toxicity , Male , Female , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Pesticides/toxicity , Sex Factors , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology
19.
Biomolecules ; 14(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39062470

ABSTRACT

Aggregation of the protein α-Synuclein (αSyn) is a hallmark of Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple systems atrophy, and alleviating the extent of αSyn pathology is an attractive strategy against neurodegeneration. The engineered binding protein ß-wrapin AS69 binds monomeric αSyn. AS69 reduces primary and secondary nucleation as well as fibril elongation in vitro. It also mitigates aSyn pathology in a mouse model based on intrastriatal injection of aSyn pre-formed fibrils (PFFs). Since the PFF-based model does not represent all aspects of PD, we tested here whether AS69 can reduce neurodegeneration resulting from αSyn overexpression. Human A53T-αSyn was overexpressed in the mouse Substantia nigra (SN) by using recombinant adeno-associated viral vector (rAAV). AS69 was also expressed by rAAV transduction. Behavioral tests and immunofluorescence staining were used as outcomes. Transduction with rAAV-αSyn resulted in αSyn pathology as reported by phospho-αSyn staining and caused degeneration of dopaminergic neurons in the SN. The co-expression of rAAV-AS69 did not reduce αSyn pathology or the degeneration of dopaminergic neurons. We conclude that αSyn monomer binding by rAAV-AS69 was insufficient to protect from aSyn pathology resulting from αSyn overexpression.


Subject(s)
Disease Models, Animal , Substantia Nigra , alpha-Synuclein , Animals , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Mice , Humans , Substantia Nigra/metabolism , Substantia Nigra/pathology , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Dependovirus/genetics , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Male , Mice, Inbred C57BL
20.
Sci Rep ; 14(1): 15107, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956066

ABSTRACT

Ferroptosis is an iron-dependent cell death form characterized by reactive oxygen species (ROS) overgeneration and lipid peroxidation. Myricetin, a flavonoid that exists in numerous plants, exhibits potent antioxidant capacity. Given that iron accumulation and ROS-provoked dopaminergic neuron death are the two main pathological hallmarks of Parkinson's disease (PD), we aimed to investigate whether myricetin decreases neuronal death through suppressing ferroptosis. The PD models were established by intraperitoneally injecting 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) into rats and by treating SH-SY5Y cells with 1-methyl-4-phenylpyridinium (MPP+), respectively. Ferroptosis was identified by assessing the levels of Fe2+, ROS, malondialdehyde (MDA), and glutathione (GSH). The results demonstrated that myricetin treatment effectively mitigated MPTP-triggered motor impairment, dopamine neuronal death, and α-synuclein (α-Syn) accumulation in PD models. Myricetin also alleviated MPTP-induced ferroptosis, as evidenced by decreased levels of Fe2+, ROS, and MDA and increased levels of GSH in the substantia nigra (SN) and serum in PD models. All these changes were reversed by erastin, a ferroptosis activator. In vitro, myricetin treatment restored SH-SY5Y cell viability and alleviated MPP+-induced SH-SY5Y cell ferroptosis. Mechanistically, myricetin accelerated nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) and subsequent glutathione peroxidase 4 (Gpx4) expression in MPP+-treated SH-SY5Y cells, two critical inhibitors of ferroptosis. Collectively, these data demonstrate that myricetin may be a potential agent for decreasing dopaminergic neuron death by inhibiting ferroptosis in PD.


Subject(s)
Disease Models, Animal , Dopaminergic Neurons , Ferroptosis , Flavonoids , Reactive Oxygen Species , Ferroptosis/drug effects , Animals , Flavonoids/pharmacology , Rats , Male , Reactive Oxygen Species/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Humans , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Cell Line, Tumor , Iron/metabolism , alpha-Synuclein/metabolism , Rats, Sprague-Dawley , Glutathione/metabolism , Lipid Peroxidation/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , NF-E2-Related Factor 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL