Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Cells ; 10(12)2021 12 02.
Article in English | MEDLINE | ID: mdl-34943913

ABSTRACT

Dendritic spines are small, thin, hair-like protrusions found on the dendritic processes of neurons. They serve as independent compartments providing large amplitudes of Ca2+ signals to achieve synaptic plasticity, provide sites for newer synapses, facilitate learning and memory. One of the common and severe complication of neurodegenerative disease is cognitive impairment, which is said to be closely associated with spine pathologies viz., decreased in spine density, spine length, spine volume, spine size etc. Many treatments targeting neurological diseases have shown to improve the spine structure and distribution. However, concise data on the various modulators of dendritic spines are imperative and a need of the hour. Hence, in this review we made an attempt to consolidate the effects of various pharmacological (cholinergic, glutamatergic, GABAergic, serotonergic, adrenergic, and dopaminergic agents) and non-pharmacological modulators (dietary interventions, enriched environment, yoga and meditation) on dendritic spines structure and functions. These data suggest that both the pharmacological and non-pharmacological modulators produced significant improvement in dendritic spine structure and functions and in turn reversing the pathologies underlying neurodegeneration. Intriguingly, the non-pharmacological approaches have shown to improve intellectual performances both in preclinical and clinical platforms, but still more technology-based evidence needs to be studied. Thus, we conclude that a combination of pharmacological and non-pharmacological intervention may restore cognitive performance synergistically via improving dendritic spine number and functions in various neurological disorders.


Subject(s)
Dendritic Spines/drug effects , Diet , Neurodegenerative Diseases/diet therapy , Neurodegenerative Diseases/drug therapy , Cholinergic Agents/therapeutic use , Cognitive Dysfunction/diet therapy , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/epidemiology , Cognitive Dysfunction/psychology , Dendritic Spines/pathology , Dendritic Spines/physiology , Excitatory Amino Acid Agents/therapeutic use , GABA Agents/therapeutic use , Humans , Meditation/psychology , Neurodegenerative Diseases/epidemiology , Neurodegenerative Diseases/psychology , Neurons/drug effects , Neurons/physiology , Synapses/drug effects , Synapses/metabolism , Yoga/psychology
2.
Mol Brain ; 14(1): 84, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34034796

ABSTRACT

Down syndrome (DS) is the most frequent genetic cause of intellectual disability including hippocampal-dependent memory deficits. We have previously reported hippocampal mTOR (mammalian target of rapamycin) hyperactivation, and related plasticity as well as memory deficits in Ts1Cje mice, a DS experimental model. Here we characterize the proteome of hippocampal synaptoneurosomes (SNs) from these mice, and found a predicted alteration of synaptic plasticity pathways, including long term depression (LTD). Accordingly, mGluR-LTD (metabotropic Glutamate Receptor-LTD) is enhanced in the hippocampus of Ts1Cje mice and this is correlated with an increased proportion of a particular category of mushroom spines in hippocampal pyramidal neurons. Remarkably, prenatal treatment of these mice with rapamycin has a positive pharmacological effect on both phenotypes, supporting the therapeutic potential of rapamycin/rapalogs for DS intellectual disability.


Subject(s)
Dendritic Spines/metabolism , Dendritic Spines/pathology , Down Syndrome/pathology , Down Syndrome/physiopathology , Long-Term Synaptic Depression , Receptors, Metabotropic Glutamate/metabolism , Sirolimus/pharmacology , Animals , Dendritic Spines/drug effects , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Long-Term Synaptic Depression/drug effects , Mice, Transgenic , Mitochondrial Proteins/metabolism , Neuronal Plasticity/drug effects , Proteomics , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Synapses/drug effects , Synapses/metabolism
3.
Chem Biol Interact ; 342: 109476, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33872575

ABSTRACT

Ketamine is the prototype for glutamate-based fast-acting antidepressants. The establishment of ketamine-like drugs is still a challenge and ascorbic acid has emerged as a candidate. This study investigated the ascorbic acid's ability to induce a fast antidepressant-like response and to improve hippocampal synaptic markers in mice subjected to chronic corticosterone (CORT) administration. CORT was administered for 21 days, followed by a single administration of ascorbic acid (1 mg ∕Kg, p.o.), ketamine (1 mg ∕Kg, i.p.) or fluoxetine (10 mg ∕Kg, p.o.) in mice. Depressive-like behavior, hippocampal synaptic proteins immunocontent, dendrite spines density in the dentate gyrus (DG) were analyzed 24 h following treatments. The administration of ascorbic acid or ketamine, but not fluoxetine, counteracted CORT-induced depressive-like behavior in the tail suspension test (TST). CORT administration reduced PSD-95, GluA1, and synapsin (synaptic markers) immunocontent, and these alterations were reversed by ascorbic acid or ketamine, but only ketamine reversed the CORT-induced reduction on GluA1 immunocontent. In the ventral and dorsal DG, CORT decreased filopodia-, thin- and stubby-shaped spines, while ascorbic acid and ketamine abolished this alteration only in filopodia spines. Ascorbic acid and ketamine increased mushroom-shaped spines density in ventral and dorsal DG. Therefore, the results show that a single administration of ascorbic acid, in a way similar to ketamine, rapidly elicits an antidepressant-like response and reverses hippocampal synaptic deficits caused by CORT, an effect associated with increased levels of synaptic proteins and dendritic remodeling.


Subject(s)
Antidepressive Agents/therapeutic use , Ascorbic Acid/therapeutic use , Depression/drug therapy , Hippocampus/drug effects , Animals , Corticosterone , Dendritic Spines/drug effects , Depression/chemically induced , Female , Hindlimb Suspension , Ketamine/therapeutic use , Mice , Neuroprotective Agents/therapeutic use
4.
Aging (Albany NY) ; 13(7): 9522-9541, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33539323

ABSTRACT

Chronic cerebral hypoperfusion (CCH) may lead to the cognitive dysfunction, but the underlying mechanisms are unclear. EGB761, extracted from Ginkgo biloba and as a phytomedicine widely used in the world, has been showed to have various neuroprotective roles and mechanisms, and therapeutic effects in Alzheimer's disease and other cognitive dysfunctions. However, improvements in cognitive function after CCH, following treatment with EGB761, have not been ascertained yet. In this study, we used the behavior test, electrophysiology, neurobiochemistry, and immunohistochemistry to investigate the EGB761's effect on CCH-induced cognitive dysfunction and identify its underlying mechanisms. The results showed that EGB761 ameliorates spatial cognitive dysfunction occurring after CCH. It may also improve impairment of the long-term potentiation, field excitable potential, synaptic transmission, and the transmission synchronization of neural circuit signals between the entorhinal cortex and hippocampal CA1. EGB761 may also reverse the inhibition of neural activity and the degeneration of dendritic spines and synaptic structure after CCH; it also prevents the downregulation of synaptic proteins molecules and pathways related to the formation and stability of dendritic spines structures. EGB761 may inhibit axon demyelination and ameliorate the inhibition of the mTOR signaling pathway after CCH to improve protein synthesis. In conclusion, EGB761 treatment after CCH may improve spatial cognitive function by ameliorating synaptic plasticity impairment, synapse degeneration, and axon demyelination by rectifying the inhibition of the mTOR signaling pathway.


Subject(s)
Brain Ischemia/complications , Cognitive Dysfunction/drug therapy , Ginkgo biloba , Neuronal Plasticity/drug effects , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Animals , Brain Ischemia/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Male , Maze Learning/drug effects , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/metabolism
5.
Neurobiol Learn Mem ; 179: 107409, 2021 03.
Article in English | MEDLINE | ID: mdl-33609738

ABSTRACT

Ghrelin (Gr) is an orexigenic peptide that acts via its specific receptor, GHSR-1a distributed throughout the brain, being mainly enriched in pituitary, cortex and hippocampus (Hp) modulating a variety of brain functions. Behavioral, electrophysiological and biochemical evidence indicated that Gr modulates the excitability and the synaptic plasticity in Hp. The present experiments were designed in order to extend the knowledge about the Gr effect upon structural synaptic plasticity since morphological and quantitative changes in spine density after Gr administration were analyzed "in vitro" and "in vivo". The results show that Gr administered to hippocampal cultures or stereotactically injected in vivo to Thy-1 mice increases the density of dendritic spines (DS) being the mushroom type highly increased in secondary and tertiary extensions. Spines classified as thin type were increased particularly in primary extensions. Furthermore, we show that Gr enhances selectively the expression of BDNF-mRNA species.


Subject(s)
Brain-Derived Neurotrophic Factor/drug effects , Ghrelin/pharmacology , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Pyramidal Cells/drug effects , RNA, Messenger/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Dendritic Spines/drug effects , Dendritic Spines/pathology , Hippocampus/cytology , Hippocampus/metabolism , Microscopy, Confocal , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , RNA, Messenger/metabolism , Rats
6.
Mol Pain ; 17: 1744806921997654, 2021.
Article in English | MEDLINE | ID: mdl-33626989

ABSTRACT

Neuropathic pain is a severe problem that is difficult to treat clinically. Reducing abnormal remodeling of dendritic spines/synapses and increasing the anti-inflammatory effects in the spinal cord dorsal horn are potential methods to treat this disease. Previous studies have reported that electroacupuncture (EA) could increase the pain threshold after peripheral nerve injury. However, the underlying mechanism is unclear. P2X7 receptors (P2X7R) mediate the activation of microglia and participate in the occurrence and development of neuropathic pain. We hypothesized that the effects of EA on relieving pain may be related to the downregulation of the P2X7R. Spinal nerve ligation (SNL) rats were used as a model in this experiment, and 2'(3')-O-(4-benzoyl)benzoyl ATP (BzATP) was used as a P2X7R agonist. We found that EA treatment decreased dendritic spine density, inhibited synaptic reconstruction and reduced inflammatory response, which is consistent with the decrease in P2X7R expression as well as the improved neurobehavioral performance. In contrast to the beneficial effects of EA, BzATP enhanced abnormal remodeling of dendritic spines/synapses and inflammation. Furthermore, the EA-mediated positive effects were reversed by BzATP, which is consistent with the increased P2X7R expression. These findings indicated that EA improves neuropathic pain by reducing abnormal dendritic spine/synaptic reconstruction and inflammation via suppressing P2X7R expression.


Subject(s)
Electroacupuncture , Neuralgia/metabolism , Neuralgia/therapy , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Dendritic Spines/drug effects , Inflammation Mediators/metabolism , Ligation , Male , Models, Biological , Nerve Tissue Proteins/metabolism , Neuralgia/physiopathology , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/pathology , Pain Threshold/drug effects , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/pathology , Spinal Nerves/drug effects , Spinal Nerves/pathology , Spinal Nerves/physiopathology
7.
J Chem Neuroanat ; 111: 101888, 2021 01.
Article in English | MEDLINE | ID: mdl-33212191

ABSTRACT

Koumine (KM) is a major alkaloid monomer in the traditional Chinese medicine herb Gelsemium elegans Benth that has exhibited therapeutic potential in clinical applications. However, the pharmacological toxicological mechanism of this drug has not been fully explored. The purpose of this study was to evaluate the impacts of KM administration at a therapeutic dose in offspring. On gestational day 0, mice were injected with KM once daily for 4 consecutive days. Male and female offspring were subjected to behavioral tests and neuropathological analyses from postnatal day 60. Prenatal KM exposure resulted in cognitive and memory impairments in the Morris water maze, Y-maze test, and novel object recognition test. The open field test and elevated plus maze test indicated that prenatal KM exposure induced anxiety-like behavior in offspring. Electrophysiological experiments demonstrated that KM exposure inhibited hippocampal long-term potentiation. Immunostaining for neurogenesis markers DCX and BrdU demonstrated that KM suppressed adult neurogenesis in the subgranular zone of the dentate gyrus. In addition, prenatal KM exposure induced a significant reduction in dendritic spine density in hippocampal neurons. Synaptic formation-related proteins were decreased in the KM group based on western blot. No sex differences in the effects of KM were observed. Collectively, our results indicate that prenatal KA exposure has detrimental neural effects on offspring. This study provides a preliminary preclinical toxicological assessment of the safety of KM use during pregnancy.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/drug effects , Cognitive Dysfunction/physiopathology , Indole Alkaloids/pharmacology , Maze Learning/drug effects , Memory/drug effects , Prenatal Exposure Delayed Effects/physiopathology , Animals , Dendritic Spines/drug effects , Doublecortin Protein , Female , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Mice , Neurogenesis/drug effects , Neurons/drug effects , Pregnancy
8.
Acta Neurobiol Exp (Wars) ; 80(3): 225-244, 2020.
Article in English | MEDLINE | ID: mdl-32990282

ABSTRACT

Alzheimer's disease (AD) has become the most prevalent neurodegenerative disorder. Given the pathogenesis of AD is unclear, there is currently no drug approved to halt or delay the progression of AD. Therefore, it is pressing to explore new targets and drugs for AD. In China, polyphenolic Chinese herbal medicine has been used for thousands of years in clinical application, and no toxic effects have been reported. In the present study, using D­galactose and aluminum­induced rat model, the effects of paeonol on AD were validated via the Morris water maze test, open field test, and elevated plus maze test. Neuronal morphology in frontal cortex was assessed using ImageJ's Sholl plugin and RESCONSTRUCT software. RhoA/Rock2/Limk1/cofilin1 signaling pathway­related molecules were determined by Western blotting. Cofilin1 and p­cofilin1 were analyzed by immunofluorescence. Results showed that pre­treatment with paeonol attenuated D­galactose and aluminum­induced behavioral dysfunction and AD­like pathological alterations in the frontal cortex. Accompanied by these changes were the alterations in the dendrite and dendritic spine densities, especially the mushroom­type and filopodia­type spines in the apical dendrites, as well as actin filaments. In addition, the activity and intracellular distribution of cofilin1 and the molecules RhoA/Rock2/Limk1 that regulate the signaling pathway for cofilin1 phosphorylation have also changed. Our data suggests that paeonol may be through reducing Aß levels to alleviate the loss of fibrillar actin and dendrites and dendritic spines via the Rho/Rock2/Limk1/cofilin1 signaling pathway in the frontal cortex, and ultimately improving AD­like behavior.


Subject(s)
Aluminum/pharmacology , Alzheimer Disease/metabolism , Dendritic Spines/metabolism , Galactose/pharmacology , rhoA GTP-Binding Protein/metabolism , Alzheimer Disease/pathology , Animals , Dendritic Spines/drug effects , Dendritic Spines/pathology , Hippocampus/drug effects , Lim Kinases/drug effects , Lim Kinases/metabolism , Neurons/drug effects , Phosphorylation/drug effects , rhoA GTP-Binding Protein/drug effects
9.
Inflammopharmacology ; 28(6): 1553-1566, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32959092

ABSTRACT

We aimed to elucidate the role of cortical and hippocampal dendritic spines on neurological deficits associated with hippocampal microgliosis, hippocampal neurogenesis, and neuroinflammation in mice with cortical compact impact (CCI) injury. In the present study, we found that CCI reduced spatial memory mean latency (10 s. vs 50 s) and motor dysfunction (130 s. vs 150 s.) in mice, as determined by Morris water maze and rotarod test, respectively. Golgi staining of cortical pyramidal neurons revealed that, compared to the controls, the CCI group treated with vehicle solution had significantly lower values of dendritic order (or dendritic branch number) (4.0 vs 6.2), total spine length (400 µm vs 620 µm) and spine density (40 spines/µm vs 60 spines/µm), but had significantly higher values of dendritic beading (40 beadings/mm vs 20 beadings/mm). Additionally, Sholl analysis showed that, compared to controls, the CCI + NS group mice had significantly lower values of dendritic intersections (1.0 vs 2.0). Immunofluorescence assay also revealed that, compared to controls, the CCI + NS group mice had significantly higher values of the newly formed hippocampal cells (1250/mm2 vs 1000/mm2) but significantly lower values of dendritic order (2.0 branch # vs 4.2 branch #), total spine length (180 µm vs 320 µm) and intersection (1.0 vs 3.0). The CCI + NS group mice further showed significantly higher numbers of microglia in the dentate gyrus of the hippocampus and higher concentrations of pro-inflammatory cytokines in the cerebrospinal fluids. All the CCI-induced spatial memory (40 s) and motor (150 s) dysfunction, deranged dendritic and spine morphology of cortical pyramidal neurons or hippocampal newly formed cells, hippocampal microgliosis, and central neuroinflammation were all significantly reduced by melatonin administration during post-CCI. Simultaneously, melatonin therapy caused an enhancement in the compensatory hippocampal neurogenesis and neurotrophic growth factors (e.g., doublecortin-1) and compensatory central anti-inflammatory cytokines. Our results indicate that melatonin attenuates the spatial memory and motor deficits via the modification of cortical and hippocampal dendritic spine morphology, hippocampal microgliosis and neurogenesis, and neuroinflammation in mice with traumatic brain injury.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Dendritic Spines/drug effects , Hippocampus/drug effects , Melatonin/pharmacology , Motor Cortex/drug effects , Neurons/drug effects , Spatial Memory/drug effects , Animals , Disease Models, Animal , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL
10.
Exp Neurol ; 333: 113413, 2020 11.
Article in English | MEDLINE | ID: mdl-32717354

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease. Pharmacotherapy with L-DOPA remains the gold-standard therapy for PD, but is often limited by the development of the common side effect of L-DOPA-induced dyskinesia (LID), which can become debilitating. The only effective treatment for disabling dyskinesia is surgical therapy (neuromodulation or lesioning), therefore effective pharmacological treatment of LID is a critical unmet need. Here, we show that sub-anesthetic doses of ketamine attenuate the development of LID in a rodent model, while also having acute anti-parkinsonian activity. The long-term anti-dyskinetic effect is mediated by brain-derived neurotrophic factor-release in the striatum, followed by activation of ERK1/2 and mTOR pathway signaling. This ultimately leads to morphological changes in dendritic spines on striatal medium spiny neurons that correlate with the behavioral effects, specifically a reduction in the density of mushroom spines, a dendritic spine phenotype that shows a high correlation with LID. These molecular and cellular changes match those occurring in hippocampus and cortex after effective sub-anesthetic ketamine treatment in preclinical models of depression, and point to common mechanisms underlying the therapeutic efficacy of ketamine for these two disorders. These preclinical mechanistic studies complement current ongoing clinical testing of sub-anesthetic ketamine for the treatment of LID by our group, and provide further evidence in support of repurposing ketamine to treat individuals with PD. Given its clinically proven therapeutic benefit for both treatment-resistant depression and several pain states, very common co-morbidities in PD, sub-anesthetic ketamine could provide multiple therapeutic benefits for PD in the future.


Subject(s)
Anesthetics, Dissociative/therapeutic use , Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Ketamine/therapeutic use , Levodopa/adverse effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Dendritic Spines/drug effects , Dendritic Spines/pathology , Depression/drug therapy , Depression/psychology , Drug Repositioning , MAP Kinase Signaling System/drug effects , Male , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/drug effects
11.
Pharmacol Res ; 158: 104863, 2020 08.
Article in English | MEDLINE | ID: mdl-32407957

ABSTRACT

Neural stem cell (NSC) neuronal differentiation requires a metabolic shift towards oxidative phosphorylation. We now show that a branched-chain amino acids-driven, persistent metabolic shift toward energy metabolism is required for full neuronal maturation. We increased energy metabolism of differentiating neurons derived both from murine NSCs and human induced pluripotent stem cells (iPSCs) by supplementing the cell culture medium with a mixture composed of branched-chain amino acids, essential amino acids, TCA cycle precursors and co-factors. We found that treated differentiating neuronal cells with enhanced energy metabolism increased: i) total dendritic length; ii) the mean number of branches and iii) the number and maturation of the dendritic spines. Furthermore, neuronal spines in treated neurons appeared more stable with stubby and mushroom phenotype and with increased expression of molecules involved in synapse formation. Treated neurons modified their mitochondrial dynamics increasing the mitochondrial fusion and, consistently with the increase of cellular ATP content, they activated cellular mTORC1 dependent p70S6 K1 anabolism. Global transcriptomic analysis further revealed that treated neurons induce Nrf2 mediated gene expression. This was correlated with a functional increase in the Reactive Oxygen Species (ROS) scavenging mechanisms. In conclusion, persistent branched-chain amino acids-driven metabolic shift toward energy metabolism enhanced neuronal differentiation and antioxidant defences. These findings offer new opportunities to pharmacologically modulate NSC neuronal differentiation and to develop effective strategies for treating neurodegenerative diseases.


Subject(s)
Amino Acids, Branched-Chain/pharmacology , Cell Differentiation/physiology , Energy Metabolism/drug effects , Neural Stem Cells/physiology , Adenosine Triphosphate/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dendritic Spines/drug effects , Dendritic Spines/ultrastructure , Humans , Induced Pluripotent Stem Cells/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Reactive Oxygen Species/metabolism , Synapses/genetics , Synapses/physiology , Synapses/ultrastructure , Transcriptome
12.
Neuromolecular Med ; 22(3): 341-358, 2020 09.
Article in English | MEDLINE | ID: mdl-32048142

ABSTRACT

Alzheimer's disease (AD) is a common neurodegenerative disease that is always accompanied by synaptic loss in the brain. Safflower yellow (SY) is the extract of safflower, a traditional Chinese medicine, which has shown neuroprotective effects in recent studies. However, the mechanism of SY in protecting synapses remains unclear. In this study, we are going to study the mechanism of how SY treats AD in terms of synaptic plasticity. We found, via behavioral experiments, that SY treatment could improve the abilities of learning and memory in APP/PS1 mice. In addition, using Golgi staining and HE staining, we found that SY treatment could reduce the loss of dendritic spines in the pathological condition and could maintain the normal physiological state of the cells in cortex and in hippocampus. In addition, the results of immunofluorescence staining and western blotting showed that SY treatment could significantly increase the expression of synapse-related proteins. Moreover, after being treated with SY, the expression of iNOS (marker of M1 microglia) declined remarkably, and the level of Arginase-1 (marker of M2 microglia) increased significantly. Finally, we found BDNF/TrkB/ERK signaling cascade was activated. These results indicate that SY enhances synaptic plasticity in APP/PS1 mice by regulating microglia activation phenotypes and BDNF/TrkB/ERK signaling pathway.


Subject(s)
Alzheimer Disease/drug therapy , Brain-Derived Neurotrophic Factor/physiology , Chalcone/analogs & derivatives , Drugs, Chinese Herbal/therapeutic use , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/physiology , Microglia/drug effects , Neuronal Plasticity/drug effects , Phytotherapy , Protein-Tyrosine Kinases/physiology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Arginase/biosynthesis , Arginase/genetics , Cerebral Cortex/chemistry , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Chalcone/therapeutic use , Dendritic Spines/drug effects , Dendritic Spines/ultrastructure , Disease Models, Animal , Donepezil/pharmacology , Donepezil/therapeutic use , Enzyme Induction/drug effects , Escape Reaction/drug effects , Female , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/pathology , Male , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/physiology , Morris Water Maze Test/drug effects , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuronal Plasticity/physiology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Presenilin-1/genetics , Random Allocation
13.
Cereb Cortex ; 29(12): 4932-4947, 2019 12 17.
Article in English | MEDLINE | ID: mdl-30877788

ABSTRACT

Preterm-born children suffer from neurological and behavioral disorders. Herein, we hypothesized that premature birth and non-maternal care of preterm newborns might disrupt neurobehavioral function, hippocampal dendritic arborization, and dendritic spine density. Additionally, we assessed whether 17ß-estradiol (E2) replacement or the TrkB receptor agonist, 7,8-dihydroxyflavone (DHF), would reverse compromised dendritic development and cognitive function in preterm newborns. These hypotheses were tested by comparing preterm (E28.5) rabbit kits cared and gavage-fed by laboratory personnel and term-kits reared and breast-fed by their mother doe at an equivalent postconceptional age. Neurobehavioral tests showed that both premature-birth and formula-feeding with non-maternal care led to increased anxiety behavior, poor social interaction, and lack of novelty preference compared with term-kits. Dendritic branching and number of total or mushroom dendritic spines were reduced in the CA1 field of preterm-kits compared with term controls. While CDC42 and Rac1/2/3 expression levels were lower, RhoA-activity was higher in preterm-kits compared with term controls. Both E2 and DHF treatment reversed prematurity-induced reduction in spine density, reduced total RhoA-GTPase levels, and enhanced cognitive function. Hence, prematurity and non-maternal care result in cognitive deficits, and reduced dendritic arbors and spines in CA1. E2 replacement or DHF treatment might reverse changes in dendritic spines and improve neurodevelopment in premature infants.


Subject(s)
Cognition/physiology , Dendritic Spines/pathology , Estradiol/pharmacology , Hippocampus/pathology , Premature Birth/physiopathology , Receptor, trkB/agonists , Animals , Cognition/drug effects , Dendritic Spines/drug effects , Estrogens/pharmacology , Female , Flavones/pharmacology , Hippocampus/drug effects , Maternal Deprivation , Pregnancy , Premature Birth/pathology , Rabbits , Receptor, trkB/drug effects
14.
Mol Pharmacol ; 95(4): 337-348, 2019 04.
Article in English | MEDLINE | ID: mdl-30696719

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder that is the major cause of dementia in the elderly. There is no cure against AD. We have recently discovered a novel transient receptor potential canonical 6 (TRPC6)-mediated intracellular signaling pathway that regulates the stability of dendritic spines and plays a role in memory formation. We have previously shown that TRPC6 agonists exert beneficial effects in models of AD and may serve as lead compounds for development of AD therapeutic agents. In the current study, we used the Clarivate Analytics Integrity database to search for additional TRPC6 agonists. We selected four compounds to study as potential neuroprotective agents. We applied bioinformatics analyses to test the basic pharmacological properties of the selected compounds. We performed in vitro screening of these compounds to validate their ability to protect mushroom spines from amyloid toxicity and determined that two of these compounds exert neuroprotective effects in the nanomolar concentration range. We have chosen one of these compounds [piperazine (PPZ)] for further testing. In agreement with previously published data, we have shown that PPZ potentiates TRPC6 channels. We demonstrated that the neuroprotective mechanism of the investigated PPZ is based on activation of neuronal store-operated calcium entry in spines. We have shown that PPZ restores long-term potentiation induction in 6-month-old 5xFAD mouse hippocampal slices. The obtained results suggest that PPZ and its derivatives are potential lead molecules for development of AD therapeutic agents.


Subject(s)
Alzheimer Disease/drug therapy , Piperazines/pharmacology , Alzheimer Disease/metabolism , Animals , Calcium Signaling/drug effects , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , TRPC Cation Channels/metabolism
15.
Mol Neurobiol ; 56(8): 5626-5642, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30659419

ABSTRACT

Chemotherapy-induced cognitive impairment, also known as "chemobrain," is a common side effect. The purpose of this study was to examine whether ginsenoside Rg1, a ginseng-derived compound, could prevent chemobrain and its underlying mechanisms. A mouse model of chemobrain was developed with three injections of docetaxel, adriamycin, and cyclophosphamide (DAC) in combination at a 2-day interval. Rg1 (5 and 10 mg/kg daily) was given 1 week prior to DAC regimen for 3 weeks. An amount of 10 mg/kg Rg1 significantly improved chemobrain-like behavior in water maze test. In vivo neuroimaging revealed that Rg1 co-treatment reversed DAC-induced decreases in prefrontal and hippocampal neuronal activity and ameliorated cortical neuronal dendritic spine elimination. It normalized DAC-caused abnormalities in the expression of multiple neuroplasticity biomarkers in the two brain regions. Rg1 suppressed DAC-induced elevation of the proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), but increased levels of the anti-inflammatory cytokines IL-4 and IL-10 in multiple sera and brain tissues. Rg1 also modulated cytokine mediators and inhibited DAC-induced microglial polarization from M2 to M1 phenotypes. In in vitro experiments, while impaired viability of PC12 neuroblastic cells and hyperactivation of BV-2 microglial cells, a model of neuroinflammation, were observed in the presence of DAC, Rg1 co-treatment strikingly reduced DAC's neurotoxic effects and neuroinflammatory response. These results indicate that Rg1 exerts its anti-chemobrain effect in an association with the inhibition of neuroinflammation by modulating microglia-mediated cytokines and the related upstream mediators, protecting neuronal activity and promoting neuroplasticity in particular brain regions associated with cognition processing.


Subject(s)
Antineoplastic Agents/adverse effects , Brain/pathology , Cognitive Dysfunction/prevention & control , Cytokines/metabolism , Ginsenosides/therapeutic use , Inflammation/drug therapy , Microglia/pathology , Neuronal Plasticity , Animals , Anxiety/complications , Anxiety/physiopathology , Behavior, Animal , Biomarkers/blood , Brain/drug effects , Brain/physiopathology , Cognition/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Cytokines/blood , Dendritic Spines/drug effects , Dendritic Spines/pathology , Female , Ginsenosides/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Inflammation/blood , Inflammation/complications , Inflammation/pathology , Locomotion/drug effects , Magnetic Resonance Imaging , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Neuronal Plasticity/drug effects , PC12 Cells , Rats
16.
Mol Neurobiol ; 56(6): 3882-3896, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30220058

ABSTRACT

Rett syndrome (RTT) is a postnatal neurodevelopmental disorder that primarily affects girls, with 95% of RTT cases resulting from mutations in the methyl-CpG-binding protein 2 (MECP2) gene. Choline, a dietary micronutrient found in most foods, has been shown to be important for brain development and function. However, the exact effects and mechanisms are still unknown. We found that 13 mg/day (1.7 × required daily intake) of postnatal choline treatment to Mecp2-conditional knockout mice rescued not only deficits in motor coordination, but also their anxiety-like behaviour and reduced social preference. Cortical neurons in the brains of Mecp2-conditional knockout mice supplemented with choline showed enhanced neuronal morphology and increased density of dendritic spines. Modelling RTT in vitro by knocking down the expression of the MeCP2 protein with shRNA, we found that choline supplementation to MeCP2-knockdown neurons increased their soma sizes and the complexity of their dendritic arbors. Rescue of the morphological defects could lead to enhanced neurotransmission, as suggested by an observed trend of increased expression of synaptic proteins and restored miniature excitatory postsynaptic current frequency in choline-supplemented MeCP2-knockdown neurons. Through the use of specific inhibitors targeting each of the known physiological pathways of choline, synthesis of phosphatidylcholine from choline was found to be essential in bringing about the changes seen in the choline-supplemented MeCP2-knockdown neurons. Taken together, these data reveal a role of choline in modulating neuronal plasticity, possibly leading to behavioural changes, and hence, a potential for using choline to treat RTT.


Subject(s)
Behavior, Animal/drug effects , Choline/pharmacology , Neuronal Plasticity/drug effects , Rett Syndrome/physiopathology , Animals , Cerebral Cortex/pathology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Dietary Supplements , Disease Models, Animal , Female , Methyl-CpG-Binding Protein 2/metabolism , Mice, Knockout , Neurites/drug effects , Neurites/metabolism , Phosphatidylcholines/biosynthesis , Rats, Sprague-Dawley
17.
Dev Neurobiol ; 78(9): 859-872, 2018 09.
Article in English | MEDLINE | ID: mdl-30022611

ABSTRACT

The cyclic nucleotide cGMP is an intracellular second messenger with important roles in neuronal functions and animals' behaviors. The phosphodiesterases (PDEs) are a family of enzymes that hydrolyze the second messengers cGMP and cAMP. Inhibition of phosphodiesterase 9 (PDE9), a main isoform of PDEs hydrolyzing cGMP, has been shown to improve learning and memory as well as cognitive function in rodents. However, the role of PDE9 in regulating neuronal structure and function in vivo remains unclear. Here we used in vivo two-photon microscopy to investigate the effect of a selective PDE9 inhibitor PF-04449613 on the activity and plasticity of dendritic spines of layer V pyramidal neurons in the mouse primary motor cortex. We found that administration of PF-04449613 increased calcium activity of dendrites and dendritic spines of layer V pyramidal neurons in mice under resting and running conditions. Chronic treatment of PF-04449613 over weeks increased dendritic spine formation and elimination under basal conditions. Furthermore, PF-04449613 treatment over 1-7 days increased the formation and survival of new spines as well as performance improvement after rotarod motor training. Taken together, our studies suggest that elevating the level of cGMP with the PDE9 inhibitor PF-04449613 increases synaptic calcium activity and learning-dependent synaptic plasticity, thereby contributing to performance improvement after learning. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 00: 000-000, 2018.


Subject(s)
Benzimidazoles/pharmacology , Dendritic Spines/drug effects , Learning/drug effects , Neuronal Plasticity/drug effects , Phenylurea Compounds/pharmacology , Pyramidal Cells/drug effects , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Cognition/drug effects , Dendrites/physiology , Dendritic Spines/physiology , Learning/physiology , Mice , Motor Cortex/drug effects , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , Synapses/drug effects , Synapses/physiology
18.
Neurosci Lett ; 675: 48-53, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29601832

ABSTRACT

Given the importance of depression and the adverse effects of conventional treatment, it is necessary to seek complementary therapies. In a rat model of depression, this study aimed to assess the behavioral and morphological effects of embedding absorbable thread in acupoints (acu-catgut), and compare the results to those of fluoxetine treatment and the corresponding control groups. Therefore, depressive-like behavior was evaluated with the forced swimming test, and dendritic morphology (in the CA1 hippocampal region) with the Golgi-Cox technique and Sholl analysis. After weaning, male Sprague-Dawley rats were housed in social isolation for 8 weeks to induce depressive-like behavior. They were then given a 21-day treatment by stimulating acupoints with acu-catgut (AC) or fluoxetine (FX) (2 mg/kg). Rats were divided into six groups: Control (socially housed), social isolation (SI), SI + AC, SI + Sham (sham embedding of thread), SI + FX and SI + VH (vehicle). Compared to fluoxetine, acu-catgut treatment was more effective in reversing depressive-like behavior elicited by SI. The SI-induced reduction in dendritic length and spine density in hippocampal CA1 pyramidal neurons was attenuated after prolonged treatment with acu-catgut or fluoxetine. Hence, both treatments proved capable of reversing depressive-like alterations caused by SI, likely due to dendritic remodeling in the hippocampus.


Subject(s)
Acupuncture Points , Antidepressive Agents, Second-Generation/administration & dosage , CA1 Region, Hippocampal/pathology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Depression/prevention & control , Fluoxetine/administration & dosage , Social Isolation , Animals , CA1 Region, Hippocampal/drug effects , Male , Physical Stimulation , Rats, Sprague-Dawley
19.
Neurosci Lett ; 646: 24-29, 2017 04 12.
Article in English | MEDLINE | ID: mdl-28279707

ABSTRACT

The medicinal plant Centella asiatica has long been used to improve memory and cognitive function. We have previously shown that a water extract from the plant (CAW) is neuroprotective against the deleterious cognitive effects of amyloid-ß (Aß) exposure in a mouse model of Alzheimer's disease, and improves learning and memory in healthy aged mice as well. This study explores the physiological underpinnings of those effects by examining how CAW, as well as chemical compounds found within the extract, modulate synaptic health in Aß-exposed neurons. Hippocampal neurons from amyloid precursor protein over-expressing Tg2576 mice and their wild-type (WT) littermates were used to investigate the effect of CAW and various compounds found within the extract on Aß-induced dendritic simplification and synaptic loss. CAW enhanced arborization and spine densities in WT neurons and prevented the diminished outgrowth of dendrites and loss of spines caused by Aß exposure in Tg2576 neurons. Triterpene compounds present in CAW were found to similarly improve arborization although they did not affect spine density. In contrast caffeoylquinic acid (CQA) compounds from CAW were able to modulate both of these endpoints, although there was specificity as to which CQAs mediated which effect. These data suggest that CAW, and several of the compounds found therein, can improve dendritic arborization and synaptic differentiation in the context of Aß exposure which may underlie the cognitive improvement observed in response to the extract in vivo. Additionally, since CAW, and its constituent compounds, also improved these endpoints in WT neurons, these results may point to a broader therapeutic utility of the extract beyond Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/pharmacology , Centella , Dendritic Spines/drug effects , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Centella/metabolism , Dendritic Spines/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Memory/drug effects , Memory/physiology , Mice, Transgenic
20.
Neuropharmacology ; 113(Pt A): 434-444, 2017 02.
Article in English | MEDLINE | ID: mdl-27773645

ABSTRACT

DMSO has a widespread use as a vehicle for water-insoluble therapeutic drug candidates but may also exert disease-relevant pharmacological effects by itself. However, its influence on the CNS has hardly been addressed. Here we examined the brain structure and function following chronic exposure to low DMSO dose at a paradigm with flawed synaptic connectivity in a preclinical transgenic mouse model for Alzheimer's disease (APPSDL mice). DMSO treatment increased spine density in a region-specific manner in the hippocampus of APPSDL mice ex vivo and in vivo. Moreover, DMSO exhibited clear influence on the behavior of this mouse line by enhancing hippocampal-dependent spatial memory accuracy, modulating hippocampal-independent olfactory habituation and displaying anxiolytic effect. Despite that most of the action of DMSO was observed in animals with elevated Aß levels, the drug did not exert its function via decreasing the oligomeric Aß species. However, challenging organotypic hippocampal slice cultures with NMDA receptor antagonist MK-801 recapitulated the effect of DMSO on spine density, indicating a tuning influence of DMSO on receptor signalization. Our findings demonstrate that DMSO should be considered as a true bioactive compound, which has the potential to be a beneficial adjuvant to counteract Aß-mediated synaptotoxicity and behavioral impairment.


Subject(s)
Alzheimer Disease/drug therapy , Dendritic Spines/drug effects , Dimethyl Sulfoxide/therapeutic use , Disease Models, Animal , Hippocampus/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Dendritic Spines/pathology , Dimethyl Sulfoxide/pharmacology , Drug Evaluation, Preclinical/methods , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Hippocampus/pathology , Male , Mice , Mice, Transgenic , Organ Culture Techniques , Spatial Memory/drug effects , Spatial Memory/physiology
SELECTION OF CITATIONS
SEARCH DETAIL