Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 367
1.
Neurochem Int ; 169: 105571, 2023 10.
Article En | MEDLINE | ID: mdl-37451345

Antidepressants are used to treat depression and some anxiety disorders, including use in pregnant patients. The pharmacological actions of these drugs generally determine the uptake and metabolism of a series of neurotransmitters, such as serotonin, norepinephrine, or dopamine, along with an increase in BDNF expression. However, many aspects of antidepressant action remain unknown, particularly whether antidepressants interfere with normal neurodevelopment when taken by pregnant women. In order to reveal cellular and molecular implications crucial to the functioning of pathways related to antidepressant effects, we performed an investigation on neuronally differentiating human SH-SY5Y cells. To our knowledge, this is the first time human SH-SY5Y cells in cultures of purely neuronal cells induced by controlled differentiation with retinoic acid are followed by short-term 48-h exposure to 0.1-10 µM escitalopram or venlafaxine. Treatment with antidepressants (1 µM) did not affect the electrophysiological properties of SH-SY5Y cells. However, the percentage of mature neurons exhibiting voltage-gated sodium currents was substantially higher in cultures pre-treated with either antidepressant. After exposure to escitalopram or venlafaxine, we observed a concentration-dependent increase in activity-dependent BDNF promoter IV activation. The assessment of neurite metrics showed significant down-regulation of neurite outgrowth upon exposure to venlafaxine. Identified changes may represent links to molecular processes of importance to depression and be involved in neurodevelopmental alterations observed in postpartum children exposed to antidepressants antenatally.


Escitalopram , Neuronal Outgrowth , Venlafaxine Hydrochloride , Child , Female , Humans , Pregnancy , Antidepressive Agents/pharmacology , Antidepressive Agents/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cell Differentiation , Cell Line, Tumor , Neuroblastoma/metabolism , Neuronal Outgrowth/drug effects , Neurons/metabolism , Venlafaxine Hydrochloride/pharmacology
2.
J Biol Chem ; 298(10): 102325, 2022 10.
Article En | MEDLINE | ID: mdl-35926710

Neurite outgrowth is an integrated whole cell response triggered by the cannabinoid-1 receptor. We sought to identify the many different biochemical pathways that contribute to this whole cell response. To understand underlying mechanisms, we identified subcellular processes (SCPs) composed of one or more biochemical pathways and their interactions required for this response. Differentially expressed genes and proteins were obtained from bulk transcriptomics and proteomic analysis of extracts from cells stimulated with a cannabinoid-1 receptor agonist. We used these differentially expressed genes and proteins to build networks of interacting SCPs by combining the expression data with prior pathway knowledge. From these SCP networks, we identified additional genes that when ablated, experimentally validated the SCP involvement in neurite outgrowth. Our experiments and informatics modeling allowed us to identify diverse SCPs such as those involved in pyrimidine metabolism, lipid biosynthesis, and mRNA splicing and stability, along with more predictable SCPs such as membrane vesicle transport and microtubule dynamics. We find that SCPs required for neurite outgrowth are widely distributed among many biochemical pathways required for constitutive cellular functions, several of which are termed 'deep', since they are distal to signaling pathways and the key SCPs directly involved in extension of the neurite. In contrast, 'proximal' SCPs are involved in microtubule growth and membrane vesicle transport dynamics required for neurite outgrowth. From these bioinformatics and dynamical models based on experimental data, we conclude that receptor-mediated regulation of subcellular functions for neurite outgrowth is both distributed, that is, involves many different biochemical pathways, and deep.


Cannabinoid Receptor Agonists , Neurites , Neuronal Outgrowth , Proteomics , Receptor, Cannabinoid, CB1 , Neurites/drug effects , Neurites/metabolism , Neuronal Outgrowth/drug effects , Signal Transduction , Receptor, Cannabinoid, CB1/metabolism , Cannabinoid Receptor Agonists/pharmacology , Humans
3.
Cell Mol Life Sci ; 79(4): 193, 2022 Mar 17.
Article En | MEDLINE | ID: mdl-35298717

Aberrant insulin-like growth factor 1 (IGF-1) signaling has been proposed as a contributing factor to the development of neurodegenerative disorders including diabetic neuropathy, and delivery of exogenous IGF-1 has been explored as a treatment for Alzheimer's disease and amyotrophic lateral sclerosis. However, the role of autocrine/paracrine IGF-1 in neuroprotection has not been well established. We therefore used in vitro cell culture systems and animal models of diabetic neuropathy to characterize endogenous IGF-1 in sensory neurons and determine the factors regulating IGF-1 expression and/or affecting neuronal health. Single-cell RNA sequencing (scRNA-Seq) and in situ hybridization analyses revealed high expression of endogenous IGF-1 in non-peptidergic neurons and satellite glial cells (SGCs) of dorsal root ganglia (DRG). Brain cortex and DRG had higher IGF-1 gene expression than sciatic nerve. Bidirectional transport of IGF-1 along sensory nerves was observed. Despite no difference in IGF-1 receptor levels, IGF-1 gene expression was significantly (P < 0.05) reduced in liver and DRG from streptozotocin (STZ)-induced type 1 diabetic rats, Zucker diabetic fatty (ZDF) rats, mice on a high-fat/ high-sugar diet and db/db type 2 diabetic mice. Hyperglycemia suppressed IGF-1 gene expression in cultured DRG neurons and this was reversed by exogenous IGF-1 or the aldose reductase inhibitor sorbinil. Transcription factors, such as NFAT1 and CEBPß, were also less enriched at the IGF-1 promoter in DRG from diabetic rats vs control rats. CEBPß overexpression promoted neurite outgrowth and mitochondrial respiration, both of which were blunted by knocking down or blocking IGF-1. Suppression of endogenous IGF-1 in diabetes may contribute to neuropathy and its upregulation at the transcriptional level by CEBPß can be a promising therapeutic approach.


Aging/metabolism , Axons/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Energy Metabolism , Insulin-Like Growth Factor I/metabolism , Sensory Receptor Cells/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Axons/drug effects , Axons/metabolism , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Respiration/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Glycolysis/drug effects , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Liver/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , NFATC Transcription Factors/metabolism , Neuronal Outgrowth/drug effects , Polymers/metabolism , Promoter Regions, Genetic/genetics , Protein Transport/drug effects , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology , Signal Transduction/drug effects
4.
Int J Mol Sci ; 23(3)2022 Feb 03.
Article En | MEDLINE | ID: mdl-35163681

The biocompatibility and the antioxidant activity of barium titanate (BaTiO3) and lithium niobate (LiNbO3) were investigated on a neuronal cell line, the PC12, to explore the possibility of using piezoelectric nanoparticles in the treatment of inner ear diseases, avoiding damage to neurons, the most delicate and sensitive human cells. The cytocompatibility of the compounds was verified by analysing cell viability, cell morphology, apoptotic markers, oxidative stress and neurite outgrowth. The results showed that BaTiO3 and LiNbO3 nanoparticles do not affect the viability, morphological features, cytochrome c distribution and production of reactive oxygen species (ROS) by PC12 cells, and stimulate neurite branching. These data suggest the biocompatibility of BaTiO3 and LiNbO3 nanoparticles, and that they could be suitable candidates to improve the efficiency of new implantable hearing devices without damaging the neuronal cells.


Antioxidants/pharmacology , Barium Compounds/pharmacology , Biocompatible Materials/pharmacology , Nanoparticles/chemistry , Neurons/drug effects , Niobium/pharmacology , Oxides/pharmacology , Titanium/pharmacology , Animals , Cell Differentiation/drug effects , Cell Shape/drug effects , Cell Survival , Cytochromes c/metabolism , Neuronal Outgrowth/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
5.
ACS Appl Mater Interfaces ; 14(5): 7230-7240, 2022 Feb 09.
Article En | MEDLINE | ID: mdl-35084814

Oriented arrays of nanofibers are ubiquitous in nature and have been widely used in recreation of the biological functions such as bone and muscle tissue regenerations. However, it remains a challenge to produce nanofiber arrays with a complex organization by using current fabrication techniques such as electrospinning and extrusion. In this work, we propose a method to fabricate the complex organization of nanofiber structures templated by a spatially varying ordered liquid crystal host, which follows the pattern produced by a maskless projection display system. By programming the synchronization of the rotated polarizer and projected segments with different shapes, various configurations of nanofiber organization ranging from a single to two-dimensional lattice of arbitrary topological defects are created in a deterministic manner. The nanofiber arrays can effectively guide and promote neurite outgrowth. The application of nanofibers with arced profiles and topological defects on neural tissue organization is also demonstrated. This finding, combined with the versatility and programmability of nanofiber structures, suggests that they will help solve challenges in nerve repair, neural regeneration, and other related tissue engineering fields.


Liquid Crystals/chemistry , Nanofibers/chemistry , Animals , Azo Compounds/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line , Cell Survival/drug effects , Mice , Nanofibers/toxicity , Nerve Regeneration/drug effects , Neuronal Outgrowth/drug effects , Neurons/cytology , Neurons/metabolism
6.
Bioorg Med Chem Lett ; 60: 128580, 2022 03 15.
Article En | MEDLINE | ID: mdl-35066142

Majucin-type Illicium sesquiterpenes with potent neurotrophic activity are considered to be promising candidates for the treatment of various neurodegenerative disease. Owing to the low-abundance metabolites in Illicium genus, there are few studies on their structural modifications, structure-activity relationships, and pharmacophoric motif. Herein, structural modifications were conducted on the hydroxyl groups at C-3 and C-6 positions of two majucin-type compounds neomajucin (1) and majucin (2), and 39 neomajucin/majucin based esters were synthesized and evaluated for their neurite outgrowth-promoting activities. Among all the target derivatives, compounds 1a, 1j, 1r, 2b, 2d, 3a, 3b, 3d and 3h displayed more potent neurite outgrowth-promoting activity than their precursors. Some interesting structure-activity relationships (SARs) were also observed. Moreover, compound 1a showed good neuroprotective effect on MPP+-induced PC12 cell damage. Finally, compounds 1a and 3a exhibited relatively no cytotoxicity to normal human H9C2 cardiac cells. This work will shed light on the development of neomajucin/majucin derivatives as potential neurotrophic agents.


Nerve Growth Factors/pharmacology , Neurodegenerative Diseases/drug therapy , Neuronal Outgrowth/drug effects , Neuroprotective Agents/pharmacology , Small Molecule Libraries/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans , Illicium/chemistry , Molecular Structure , Nerve Growth Factors/chemical synthesis , Nerve Growth Factors/chemistry , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , PC12 Cells , Rats , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
7.
Biomed Pharmacother ; 147: 112663, 2022 Mar.
Article En | MEDLINE | ID: mdl-35093759

Memory-enhancing agents have long been required for various reasons such as for obtaining a good score in a test in the young and for retaining memory in the aged. Although many studies have found that several natural products may be good candidates for memory enhancement, there is still a need for better agents. The present study investigated whether rubrofusarin, an active ingredient in Cassiae semen, enhances learning and memory in normal mice. Passive avoidance and Morris water maze tests were performed to determine the memory-enhancing ability of rubrofusarin. To investigate synaptic function, hippocampal long-term potentiation (LTP) was measured. Western blotting was performed to determine protein levels. To investigate neurite outgrowth, DCX immunohistochemistry and cell culture were utilised. Rubrofusarin (1, 3, 10, 30 mg/kg) enhanced memory in passive avoidance and Morris water maze tests. Moreover, rubrofusarin ameliorated scopolamine-induced memory impairment. In the rubrofusarin-treated group, high-frequency stimulation induced higher LTP in the hippocampal Schaffer-collateral pathway compared to the control group. The rubrofusarin-treated group showed a higher number of DCX-positive immature neurons with an increase in the length of dendrites compared to the control group in the hippocampal dentate gyrus region. In vitro experiments showed that rubrofusarin facilitated neurite outgrowth in neuro2a cells through extracellular signal-regulated kinase (ERK). Finally, we found that extracellular signal-regulated kinase (ERK) is required for rubrofusarin-induced enhancement of neurite outgrowth, learning and memory. These results demonstrate that rubrofusarin enhances learning and memory and neurite outgrowth, and these might need activation of ERK pathway.


Cognition/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Neuronal Outgrowth/drug effects , Pyrones/pharmacology , Animals , Cell Culture Techniques , Dose-Response Relationship, Drug , Hippocampus/drug effects , Learning/drug effects , Long-Term Potentiation/drug effects , Male , Memory/drug effects , Mice , Pyrones/administration & dosage
8.
Mol Neurobiol ; 59(1): 61-76, 2022 Jan.
Article En | MEDLINE | ID: mdl-34623600

Parkinson's disease (PD) is a neurodegenerative disease characterised by the progressive degeneration of midbrain dopaminergic neurons, coupled with the intracellular accumulation of α-synuclein. Axonal degeneration is a central part of the pathology of PD. While the majority of PD cases are sporadic, some are genetic; the G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is the most common genetic form. The application of neurotrophic factors to protect dopaminergic neurons is a proposed experimental therapy. One such neurotrophic factor is growth differentiation factor (GDF)5. GDF5 is a dopaminergic neurotrophic factor that has been shown to upregulate the expression of a protein called nucleoside diphosphate kinase A (NME1). However, whether NME1 is neuroprotective in cell models of axonal degeneration of relevance to PD is unknown. Here we show that treatment with NME1 can promote neurite growth in SH-SY5Y cells, and in cultured dopaminergic neurons treated with the neurotoxin 6-hydroxydopamine (6-OHDA). Similar effects of NME1 were found in SH-SY5Y cells and dopaminergic neurons overexpressing human wild-type α-synuclein, and in stable SH-SY5Y cell lines carrying the G2019S LRRK2 mutation. We found that the effects of NME1 require the RORα/ROR2 receptors. Furthermore, increased NF-κB-dependent transcription was partially required for the neurite growth-promoting effects of NME1. Finally, a combined bioinformatics and biochemical analysis of the mitochondrial oxygen consumption rate revealed that NME1 enhanced mitochondrial function, which is known to be impaired in PD. These data show that recombinant NME1 is worthy of further study as a potential therapeutic agent for axonal protection in PD.


Dopaminergic Neurons/drug effects , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , NM23 Nucleoside Diphosphate Kinases/pharmacology , Nerve Degeneration/prevention & control , Neurites/drug effects , Neuroprotective Agents/pharmacology , alpha-Synuclein/genetics , Cell Line, Tumor , Dopaminergic Neurons/pathology , Humans , Nerve Degeneration/genetics , Neurites/pathology , Neuronal Outgrowth/drug effects
9.
Nanotechnology ; 33(13)2022 Jan 07.
Article En | MEDLINE | ID: mdl-34929684

Several transport vectors, including nanoparticles, have been reported to be used for the delivery of therapeutic medicines crossing the impermeable blood-brain barrier (BBB) to treat the diseases in the central nerve system (CNS), such as traumatic brain injury (TBI). Poly(n-butyl-2-cyanoacrylate) (PBCA) nanoparticles, made from biocompatible material, are regarded as a better potential delivery tool than others such as gold nanoparticles due to their degradabilityin vivo. However, little is known whether PBCA nanoparticles can be used to deliver neurotrophic factors into the brain to treat TBI. In this study, we first synthesized PBCA-carriedß-nerve growth factor, a neurotrophic agent with a large molecular weight, and then intravenously injected the compound into TBI rats. We found that despite undergoing several synthesis steps and host circulation,ß-NGF was able to be successfully delivered into the injured brain by PBCA nanoparticles, still maintain its neurotrophic activity for neurite outgrowth, and reduce the mortality of TBI rats. Our findings indicate that PBCA nanoparticles, with Tween 80, are an efficient delivery vector and a protective reservoir for large molecular therapeutic agents to treat TBI intravenously.


Brain Injuries, Traumatic/drug therapy , Drug Delivery Systems , Enbucrilate/administration & dosage , Nanoparticles/administration & dosage , Nerve Growth Factor/administration & dosage , Neuronal Outgrowth/drug effects , Animals , Cells, Cultured , Enbucrilate/chemistry , Male , Nanoparticles/chemistry , PC12 Cells , Rats , Rats, Sprague-Dawley
10.
J Pharmacol Sci ; 148(1): 103-107, 2022 Jan.
Article En | MEDLINE | ID: mdl-34924113

We have reported that nicotine has a neurotrophic action on peripheral adrenergic nerves in vivo, which is mediated by α7 nicotinic acetylcholine receptors (nAChRs). To clarify the possible mechanisms, the present study further investigated the effect of nicotine on neurite outgrowth in tyrosine hydroxylase (TH)-positive superior cervical ganglia (SCG) cells isolated from neonatal rats in vitro. Nicotine at low concentrations (0.01-0.3 mM) increased the number of neurite outgrowths in TH-immunopositive SCG cells, while high concentrations of nicotine (1-10 mM) gradually reduced it, and only 10 mM nicotine was markedly inhibited compared to the control. A 100 µM of nicotine-induced increase in neurite numbers depended on the exposure time and was inhibited by treatment with the nAChR antagonist hexamethonium (Hex) and α7 nAChR antagonist α-bungarotoxin (α-Bgtx). The nicotine (10 mM)-induced a significant decrease in neurite outgrowth in SCG, which was perfectly canceled by Hex to the control level but not by α-Bgtx. These results suggest that nicotine has a regulatory neurotrophic action mediated by both α7 nAChR and other subtypes in TH-positive SCG cells of rats.


Nerve Growth Factors , Neurites/drug effects , Neurites/physiology , Neuronal Outgrowth/drug effects , Nicotine/pharmacology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology , Animals , Cells, Cultured , Rats , alpha7 Nicotinic Acetylcholine Receptor/physiology
11.
Toxicology ; 465: 153052, 2022 01 15.
Article En | MEDLINE | ID: mdl-34838597

Bisphenol-A (BPA), an environmental endocrine disruptor, is toxic to the central nervous system. Although recent studies have shown BPA-induced neurotoxicity, it is far from clear what precisely epigenetic mechanisms are involved in BPA-induced cognitive deficits. In this study, pheochromocytoma (PC12) cells were treated with BPA at 1 µM for 36 h in vitro. In vivo, C57BL/6 mice were administered to BPA at a dose of 1 mg/kg/day for 10 weeks. The results showed that 1 µM BPA exposure for 36 h impaired neurite outgrowth of PC12 cells through decreasing the primary and secondary branches. Besides, BPA exposure decreased the level of Ac-H3K9 (histone H3 Lys9 acetylation) by upregulating the expression of HDAC2 (histone deacetylases 2) in PC12 cells. Furthermore, treatment of both TSA (Trichostatin A, inhibitor of the histone deacetylase) and shHDAC2 plasmid (HDAC2 knockdown construct) resulted in amelioration neurite outgrowth deficits induced by BPA. In addition, it was shown that repression of HDAC2 could markedly rescue the spine density impairment in the hippocampus and prevent the cognitive impairment caused by BPA exposure in mice. Collectively, HDAC2 plays an essential role in BPA-induced neurotoxicity, which provides a potential molecular target for medical intervention.


Benzhydryl Compounds/toxicity , Dendritic Spines/drug effects , Environmental Pollutants/toxicity , Hippocampus/drug effects , Histone Deacetylase 2/metabolism , Neurites/drug effects , Neurotoxicity Syndromes/etiology , Phenols/toxicity , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Dendritic Spines/enzymology , Dendritic Spines/pathology , Female , Hippocampus/enzymology , Hippocampus/pathology , Hippocampus/physiopathology , Histone Deacetylase 2/genetics , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Neurites/enzymology , Neurites/pathology , Neuronal Outgrowth/drug effects , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/physiopathology , PC12 Cells , Rats , Up-Regulation
12.
Nutrients ; 13(12)2021 Nov 27.
Article En | MEDLINE | ID: mdl-34959841

Gongjin-dan (GJD) is a multiherbal formula produced from 10 medicinal herbs and has been traditonally used as an oriental medicine to treat cardiovascular diseases, alcoholic hepatitis, mild dementia, and anemia. Additionally, increasing evidence suggests that GJD exerts neuroprotective effects by suppressing inflammation and oxidative stress-induced events to prevent neurological diseases. However, the mechanism by which GJD prevents oxidative stress-induced neuronal injury in a mature neuron remains unknown. Here, we examined the preventive effect and mechanism of GJD on primary cortical neurons exposed to hydrogen peroxide (H2O2). In the neuroprotection signaling pathway, Sirtuin1 is involved in neuroprotective action as a therapeutic target for neurological diseases. After pre-treatment with GJD at three concentrations (10, 25, and 50 µg/mL) and stimulation by H2O2 (30 µM) for 24 h, the influence of GJD on Sirtuin1 activation was assessed using immunocytochemistry, real-time PCR, western blotting, and flow cytometry. GJD effectively ameliorated H2O2-induced neuronal death against oxidative damage through Sirtuin1 activation. In addition, GJD-induced Sirtuin1 activation accelerated elongation of new axons and formation of synapses via increased expression of nerve growth factor and brain-derived neurotrophic factor, as well as regeneration-related genes. Thus, GJD shows potential for preventing neurological diseases via Sirtuin1 activation.


Neuronal Outgrowth/drug effects , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Signal Transduction/drug effects , Sirtuin 1/metabolism , Animals , Cerebral Cortex/growth & development , Hydrogen Peroxide/adverse effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley
13.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article En | MEDLINE | ID: mdl-34830376

Fungicides often cause genotoxic stress and neurodevelopmental disorders such as autism (ASD). Fungicide-azoxystrobin (AZOX) showed acute and chronic toxicity to various organisms, and remained a concern for ill effects in developing neurons. We evaluated the neurotoxicity of AZOX in developing mouse brains, and observed prenatal exposure to AZOX reduced neuronal viability, neurite outgrowth, and cortical migration process in developing brains. The 50% inhibitory concentration (IC50) of AZOX for acute (24 h) and chronic (7 days) exposures were 30 and 10 µM, respectively. Loss in viability was due to the accumulation of reactive oxygen species (ROS), and inhibited neurite outgrowth was due to the deactivation of mTORC1 kinase activity. Pretreatment with ROS scavenger- N-acetylcysteine (NAC) reserved the viability loss and forced activation of mTORC1 kinase revived the neurite outgrowth in AZOX treated neurons. Intra-amniotic injection of AZOX coupled with in utero electroporation of GFP-labelled plasmid in E15.5 mouse was performed and 20 mg/kg AZOX inhibited radial neuronal migration. Moreover, the accumulation of mitochondria was significantly reduced in AZOX treated primary neurons, indicative of mitochondrial deactivation and induction of apoptosis, which was quantified by Bcl2/Bax ratio and caspase 3 cleavage assay. This study elucidated the neurotoxicity of AZOX and explained the possible cure from it.


Apoptosis/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Pyrimidines/pharmacology , Strobilurins/pharmacology , Acetylcysteine/pharmacology , Animals , Autistic Disorder/chemically induced , Autistic Disorder/genetics , Autistic Disorder/pathology , Cell Movement/drug effects , Cell Survival/drug effects , Female , Fungicides, Industrial/toxicity , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mitochondria/drug effects , Neuronal Outgrowth/drug effects , Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects , Pyrimidines/toxicity , Reactive Oxygen Species/antagonists & inhibitors , Strobilurins/toxicity
14.
Cells ; 10(11)2021 11 09.
Article En | MEDLINE | ID: mdl-34831318

Alzheimer's disease (AD) is a common neurodegenerative disease presenting with progressive memory and cognitive impairments. One of the pathogenic mechanisms of AD is attributed to the aggregation of misfolded amyloid ß (Aß), which induces neurotoxicity by reducing the expression of brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase B (TRKB) and increasing oxidative stress, caspase-1, and acetylcholinesterase (AChE) activities. Here, we have found the potential of two novel synthetic coumarin derivatives, ZN014 and ZN015, for the inhibition of Aß and neuroprotection in SH-SY5Y neuroblastoma cell models for AD. In SH-SY5Y cells expressing the GFP-tagged Aß-folding reporter, both ZN compounds reduced Aß aggregation, oxidative stress, activities of caspase-1 and AChE, as well as increased neurite outgrowth. By activating TRKB-mediated extracellular signal-regulated kinase (ERK) and AKT serine/threonine kinase 1 (AKT) signaling, these two ZN compounds also upregulated the cAMP-response-element binding protein (CREB) and its downstream BDNF and anti-apoptotic B-cell lymphoma 2 (BCL2). Knockdown of TRKB attenuated the neuroprotective effects of ZN014 and ZN015. A parallel artificial membrane permeability assay showed that ZN014 and ZN015 could be characterized as blood-brain barrier permeable. Our results suggest ZN014 and ZN015 as novel therapeutic candidates for AD and demonstrate that ZN014 and ZN015 reduce Aß neurotoxicity via pleiotropic mechanisms.


Amyloid beta-Peptides/toxicity , Coumarins/pharmacology , Green Fluorescent Proteins/toxicity , Neuroprotective Agents/pharmacology , Acetylcholinesterase/metabolism , Biological Availability , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Caspase 1/metabolism , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Coumarins/chemistry , Gene Knockdown Techniques , Humans , Neuronal Outgrowth/drug effects , Protein Aggregates , Reactive Oxygen Species/metabolism , Receptor, trkB/metabolism
15.
Sci Rep ; 11(1): 22161, 2021 11 12.
Article En | MEDLINE | ID: mdl-34772945

Neurodegenerative diseases are characterized by chronic neuroinflammation and may perpetuate ongoing fibrotic reactions within the central nervous system. Unfortunately, there is no therapeutic available that treats neurodegenerative inflammation and its sequelae. Here we utilize cromolyn, a mast cell inhibitor with anti-inflammatory capabilities, and its fluorinated analogue F-cromolyn to study fibrosis-related protein regulation and secretion downstream of neuroinflammation and their ability to promote microglial phagocytosis and neurite outgrowth. In this report, RNA-seq analysis shows that administration of the pro-inflammatory cytokine TNF-α to HMC3 human microglia results in a robust upregulation of fibrosis-associated genes. Subsequent treatment with cromolyn and F-cromolyn resulted in reduced secretion of collagen XVIII, fibronectin, and tenascin-c. Additionally, we show that cromolyn and F-cromolyn reduce pro-inflammatory proteins PLP1, PELP1, HSP90, IL-2, GRO-α, Eotaxin, and VEGF-Α, while promoting secretion of anti-inflammatory IL-4 in HMC3 microglia. Furthermore, cromolyn and F-cromolyn augment neurite outgrowth in PC12 neuronal cells in concert with nerve growth factor. Treatment also differentially altered secretion of neurogenesis-related proteins TTL, PROX1, Rab35, and CSDE1 in HMC3 microglia. Finally, iPSC-derived human microglia more readily phagocytose Aß42 with cromolyn and F-cromolyn relative to controls. We propose the cromolyn platform targets multiple proteins upstream of PI3K/Akt/mTOR, NF-κB, and GSK-3ß signaling pathways to affect cytokine, chemokine, and fibrosis-related protein expression.


Cromolyn Sodium/pharmacology , Microglia/immunology , Microglia/metabolism , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuronal Outgrowth/drug effects , Phagocytosis/drug effects , Phagocytosis/immunology , Amyloid beta-Peptides/metabolism , Animals , Biomarkers , Cell Line , Computational Biology/methods , Cytokines/metabolism , Disease Susceptibility , Fibrosis , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Humans , Microglia/pathology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/pathology , Peptide Fragments/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proteome , Signal Transduction/drug effects
16.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article En | MEDLINE | ID: mdl-34769111

Characterization of new pharmacological targets is a promising approach in research of neurorepair mechanisms. The G protein-coupled receptor 17 (GPR17) has recently been proposed as an interesting pharmacological target, e.g., in neuroregenerative processes. Using the well-established ex vivo model of organotypic slice co-cultures of the mesocortical dopaminergic system (prefrontal cortex (PFC) and substantia nigra/ventral tegmental area (SN/VTA) complex), the influence of GPR17 ligands on neurite outgrowth from SN/VTA to the PFC was investigated. The growth-promoting effects of Montelukast (MTK; GPR17- and cysteinyl-leukotriene receptor antagonist), the glial cell line-derived neurotrophic factor (GDNF) and of two potent, selective GPR17 agonists (PSB-16484 and PSB-16282) were characterized. Treatment with MTK resulted in a significant increase in mean neurite density, comparable with the effects of GDNF. The combination of MTK and GPR17 agonist PSB-16484 significantly inhibited neuronal growth. qPCR studies revealed an MTK-induced elevated mRNA-expression of genes relevant for neuronal growth. Immunofluorescence labelling showed a marked expression of GPR17 on NG2-positive glia. Western blot and RT-qPCR analysis of untreated cultures suggest a time-dependent, injury-induced stimulation of GPR17. In conclusion, MTK was identified as a stimulator of neurite fibre outgrowth, mediating its effects through GPR17, highlighting GPR17 as an interesting therapeutic target in neuronal regeneration.


Acetates/pharmacology , Cyclopropanes/pharmacology , Leukotriene Antagonists/pharmacology , Neuronal Outgrowth/drug effects , Quinolines/pharmacology , Receptors, G-Protein-Coupled/metabolism , Sulfides/pharmacology , Animals , Animals, Newborn , Coculture Techniques , Drug Evaluation, Preclinical , Female , Male , Nerve Regeneration/drug effects , Neuronal Outgrowth/genetics , Rats
17.
Exp Eye Res ; 212: 108797, 2021 11.
Article En | MEDLINE | ID: mdl-34687724

Increasing the level of cyclic adenosine 3, 5'-monophosphate is an important mechanism for axon outgrowth and recovery of central nervous system function. This study aimed to investigate the effects of papaverine, a non-specific phosphodiesterase inhibitor, on axon outgrowth of primary retinal ganglion cells from Sprague Dawley rats. Experiments were performed on primary retinal ganglion cells extracted from Sprague Dawley rat pups within 48-72 h of birth. At 24 h after seeding, immunofluorescence was used to identify and calculate the purity of retinal ganglion cells isolated by an improved two-step immunopanning method developed by author Sujia Ma. The effects of a range of papaverine concentrations on axon outgrowth of primary retinal ganglion cells cultures were observed by immunofluorescence and measured by ImageJ software at three different time points: 24, 48, and 72 h. The ability of papaverine to enable retinal ganglion cells to overcome the inhibitory effects of glial scar component chondroitin sulfate proteoglycans was examined using chondroitin sulfate proteoglycans-coated culture plates. Rp-adenosine 3',5'-cyclic monophosphorothioate triethylammonium salt, a blocking agent of cyclic adenosine 3, 5'-monophosphate, and dibutyryl cyclic adenosine 3, 5'-monophosphate, an analogue of cyclic adenosine 3, 5'-monophosphate, were used to explore the mechanism of papaverine in promoting retinal ganglion cells axon outgrowth. Our study shows 2 µg/mL papaverine concentration significantly promoted axon outgrowth in primary retinal ganglion cells and restored axon outgrowth of these cells on chondroitin sulfate proteoglycans. Axon outgrowth was blocked by Rp-adenosine 3',5'-cyclic monophosphorothioate triethylammonium salt and obviously promoted by dibutyryl cyclic adenosine 3, 5'-monophosphate. Our study is the first to describe the use of papaverine to promote axon outgrowth of retinal ganglion cells. These results may help to expand the application of papaverine, and they provide a cytological basis for papaverine in the treatment of optic nerve injury caused by glaucoma and other diseases.


Glaucoma/drug therapy , Nerve Regeneration/physiology , Neuronal Outgrowth/physiology , Papaverine/pharmacology , Retinal Ganglion Cells/drug effects , Animals , Cells, Cultured , Disease Models, Animal , Glaucoma/diagnosis , Nerve Regeneration/drug effects , Neuronal Outgrowth/drug effects , Phosphodiesterase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/pathology
18.
PLoS One ; 16(10): e0258289, 2021.
Article En | MEDLINE | ID: mdl-34634077

The glial cell line-derived neurotrophic factor (GDNF) is involved in the survival of dopaminergic neurons. Besides, GDNF can also induce axonal growth and creation of new functional synapses. GDNF potential is promising for translation to treat diseases associated with neuronal death: neurodegenerative disorders, ischemic stroke, and cerebral or spinal cord damages. Unproductive clinical trials of GDNF for Parkinson's disease treatment have induced to study this failure. A reason could be due to irrelevant producer cells that cannot perform the required post-translational modifications. The biological activity of recombinant mGDNF produced by E. coli have been compared with mGDNF produced by human cells HEK293. mGDNF variants were tested with PC12 cells, rat embryonic spinal ganglion cells, and SH-SY5Y human neuroblastoma cells in vitro as well as with a mouse model of the Parkinson's disease in vivo. Both in vitro and in vivo the best neuro-inductive ability belongs to mGDNF produced by HEK293 cells. Keywords: GDNF, neural differentiation, bacterial and mammalian expression systems, cell cultures, model of Parkinson's disease.


Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neurons/physiology , Recombinant Proteins/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Disease Models, Animal , Escherichia coli , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , HEK293 Cells , Humans , Mice, Inbred C57BL , Neuronal Outgrowth/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , PC12 Cells , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Rats , Recombinant Proteins/therapeutic use , Tyrosine 3-Monooxygenase/metabolism
19.
Am J Physiol Cell Physiol ; 321(5): C770-C778, 2021 11 01.
Article En | MEDLINE | ID: mdl-34495765

Skeletal muscle is an endocrine organ secreting exercise-induced factors (exerkines), which play a pivotal role in interorgan cross talk. Using mass spectrometry (MS)-based proteomics, we characterized the secretome and identified thymosin ß4 (TMSB4X) as the most upregulated secreted protein in the media of contracting C2C12 myotubes. TMSB4X was also acutely increased in the plasma of exercising humans irrespective of the insulin resistance condition or exercise mode. Treatment of mice with TMSB4X did not ameliorate the metabolic disruptions associated with diet induced-obesity, nor did it enhance muscle regeneration in vivo. However, TMSB4X increased osteoblast proliferation and neurite outgrowth, consistent with its WADA classification as a prohibited growth factor. Therefore, we report TMSB4X as a human exerkine with a potential role in cellular cross talk.


Cell Proliferation/drug effects , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Neuronal Outgrowth/drug effects , Osteoblasts/drug effects , Thymosin/metabolism , Thymosin/pharmacology , Animals , Case-Control Studies , Cell Line, Tumor , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Humans , Insulin Resistance , Male , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Osteoblasts/pathology , Physical Endurance , Proteomics , Signal Transduction , Tandem Mass Spectrometry
20.
Sci Rep ; 11(1): 18124, 2021 09 13.
Article En | MEDLINE | ID: mdl-34518601

The treatment of spinal cord injury (SCI) is a complex challenge in regenerative medicine, complicated by the low intrinsic capacity of CNS neurons to regenerate their axons and the heterogeneity in size, shape and extent of human injuries. For example, some contusion injuries do not compromise the dura mater and in such cases implantation of preformed scaffolds or drug delivery systems may cause further damage. Injectable in situ thermosensitive scaffolds are therefore a less invasive alternative. In this study, we report the development of a novel, flowable, thermosensitive, injectable drug delivery system comprising bovine collagen (BC) and fibrinogen (FB) that forms a solid BC/FB gel (Gel) immediately upon exposure to physiological conditions and can be used to deliver reparative drugs, such as the naturally occurring anti-inflammatory, anti-scarring agent Decorin, into adult rat spinal cord lesion sites. In dorsal column lesions of adult rats treated with the Gel + Decorin, cavitation was completely suppressed and instead lesion sites became filled with injury-responsive cells and extracellular matrix materials, including collagen and laminin. Decorin increased the intrinsic potential of dorsal root ganglion neurons (DRGN) by increasing their expression of regeneration associated genes (RAGs), enhanced local axon regeneration/sprouting, as evidenced both histologically and by improved electrophysiological, locomotor and sensory function recovery. These results suggest that this drug formulated, injectable hydrogel has the potential to be further studied and translated into the clinic.


Collagen/chemistry , Decorin/administration & dosage , Fibrinogen/chemistry , Hydrogels/chemistry , Spinal Cord Injuries/rehabilitation , Spinal Cord Injuries/therapy , Action Potentials/drug effects , Animals , Biomarkers , Cattle , Cell Survival/drug effects , Disease Models, Animal , Extracellular Matrix , Ganglia, Spinal , Matrix Metalloproteinases/metabolism , Nerve Regeneration/drug effects , Neuronal Outgrowth/drug effects , Neurons/physiology , Recovery of Function , Regenerative Medicine , Spinal Cord Injuries/etiology , Spinal Cord Injuries/metabolism , Wound Healing
...