Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Neurochem Res ; 46(8): 2019-2032, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34076791

ABSTRACT

G-protein coupled estrogen receptor 1 (GPER1) is a novel type of estrogen receptor. Several studies have shown that it has an anti-inflammatory action,which plays an important role in remyelination and cognitive ability adjustment. However, whether it is involved in the development of temporal lobe epilepsy (TLE) is still unknown. The present study established a TLE model by intraperitoneal injection of lithium chloride (3 mmol/kg) and pilocarpine (50 mg/kg) in rats to study the effect of GPER1 in the synaptic plasticity during the development of temporal lobe epilepsy. A microinjection cannula was implanted into the lateral ventricle region of rats via a stereotaxic instrument. G-1 is the specific GPER1 agonist and G15 is the specific GPER1 antagonist. The G1 or G15 and Dimethyl sulfoxide were injected into the rat brains in the intervention groups and control group, respectively. After G1 intervention, the learning and memory abilities and hippocampal neuron damage in epileptic rats were significantly improved, while G15 weakened the neuroprotective effect of GPER1. Meanwhile, G1 controlled the abnormal formation of hippocampal mossy fiber sprouting caused by seizures, and participated in the regulation of synaptic plasticity by reducing the expression of Synapsin I and increasing the expression of gephyrin. Inhibitory synapse gephyrin may play a significant role in synaptic plasticity.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Neuronal Plasticity/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/pathology , Hippocampus/drug effects , Hippocampus/pathology , Learning/drug effects , Lithium Chloride , Male , Membrane Proteins/metabolism , Memory/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/physiology , Pilocarpine , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Synapsins/metabolism
2.
Eur J Neurosci ; 52(9): 4100-4114, 2020 11.
Article in English | MEDLINE | ID: mdl-32588491

ABSTRACT

Sleep pressure that builds up gradually during the extended wakefulness results in sleep rebound. Several lines of evidence, however, suggest that wake per se may not be sufficient to drive sleep rebound and that rapid eye movement (REM) and non-rapid eye movement (NREM) sleep rebound may be differentially regulated. In this study, we investigated the relative contribution of brain versus physical activities in REM and NREM sleep rebound by four sets of experiments. First, we forced locomotion in rats in a rotating wheel for 4 hr and examined subsequent sleep rebound. Second, we exposed the rats lacking homeostatic sleep response after prolonged quiet wakefulness and arousal brain activity induced by chemoactivation of parabrachial nucleus to the same rotating wheel paradigm and tested if physical activity could rescue the sleep homeostasis. Third, we varied motor activity levels while concurrently inhibiting the cortical activity by administering ketamine or xylazine (motor inhibitor), or ketamine + xylazine mixture and investigated if motor activity in the absence of activated cortex can cause NREM sleep rebound. Fourth and finally, we manipulated cortical activity by administering ketamine (that induced active wakefulness and waking brain) alone or in combination with atropine (that selectively inhibits the cortex) and studied if cortical inhibition irrespective of motor activity levels can block REM sleep rebound. Our results demonstrate that motor activity but not cortical activity determines NREM sleep rebound whereas cortical activity but not motor activity determines REM sleep rebound.


Subject(s)
Electroencephalography , Sleep , Animals , Homeostasis , Rats , Sleep, REM , Wakefulness
3.
Int J Neurosci ; 130(2): 144-152, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31554446

ABSTRACT

Purpose: Recent studies have shown that growth-associated protein-43 (GAP-43) may influence the mitotic-spindle orientation of Madin-Darby Canine Kidney (MDCK) cells through interacting with G proteins in vitro. However, whether GAP-43 interacts with the G proteins under the influence of mitotic spindle positioning related to the orientation of cell division during neurogenesis remains unclear. In order to explore the molecular mechanism in vivo, the GAP-43 transgenic mice were produced and the angles of cell division in the ventricular zone (VZ) during neurogenesis (embryonic period between 13.5 and 17.5 days) were measured in both transgenic mice and wild type mice by spindle angle analysis.Materials and methods: The interaction of GAP-43 and Gαi was detected by co-immunoprecipitation (co-IP), whereas the localization of GAP-43 was determined by immunofluorescence.Results: The results obtained using co-IP and immunofluorescence showed that GAP-43 is localized on the cell membrane and interacts with Gαi. This interaction dramatically induced a significant increase in the proportion of horizontally and intermediately dividing cells during the embryonic period of 13.5 days in the transgenic mouse brain, as observed by spindle angle analysis.Conclusions: It can be concluded that GAP-43 is involved in the orientation of cell division by interacting with Gαi, and that this may be an important mechanism for neurogenesis in the mammalian brain.


Subject(s)
Brain/growth & development , Cell Division/physiology , GAP-43 Protein/physiology , GTP-Binding Protein alpha Subunits/metabolism , Neurogenesis/physiology , Animals , Brain/metabolism , Embryo, Mammalian , GAP-43 Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Cell Tissue Res ; 364(1): 17-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26463048

ABSTRACT

In order to create an optimal microenvironment for neural regeneration in the lesion area after spinal cord injury (SCI), we fabricated a novel scaffold composed of a hyaluronic acid (HA) hydrogel with a longitudinal multi-tubular conformation. The scaffold was modified by binding with an anti-Nogo receptor antibody (antiNgR) and mixed further with poly(lactic-co-glycolic acid) (PLGA) microspheres containing brain-derived neurotrophic factor and vascular endothelial growth factor (HA+PLGA). In the rat, after implantation of this composite into an injured area created by a dorsal hemisection at T9-10 of the spinal cord, favorable effects were seen with regard to the promotion of spinal repair, including excellent integration of the implants with host tissue, inhibition of inflammation, and gliosis. In particular, large numbers of new blood vessels and regenerated nerve fibers were found within and around the implants. Simultaneously, the implanted rats exhibited improved locomotor recovery. Thus, this novel composite material might provide a suitable microenvironment for neural regeneration following SCI.


Subject(s)
Hyaluronic Acid/pharmacology , Lactic Acid/pharmacology , Microspheres , Polyglycolic Acid/pharmacology , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Tissue Scaffolds , Animals , Female , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley
5.
Sleep Med ; 113: 49-55, 2024 01.
Article in English | MEDLINE | ID: mdl-37984017

ABSTRACT

INTRODUCTION: The basal forebrain (BF) and the medial septum (MS) respectively drive neuronal activity of cerebral cortex and hippocampus (HPC) in sleep-wake cycle. Our previous studies of lesions and neuronal circuit tracing have shown that the pontine parabrachial nucleus (PB) projections to the BF and MS may be a key circuit for cortical and HPC arousal. AIMS: This study aims to demonstrate that PB projections to the BF and MS activate the cerebral cortex and HPC. RESULTS: By using chemogenetic stimulation of the BF, the PB-BF and the PB-MS pathway combined with electroencephalogram (EEG) Fast Fourier Transformation (FFT) analysis in rats, we demonstrated that chemogenetic stimulation of the BF or PB neurons projecting to the BF activated the cerebral cortex while chemogenetic stimulation of the MS or PB neurons projecting to the MS activated HPC activity, in sleep and wake state. These stimulations did not significantly alter sleep-wake amounts. CONCLUSIONS: Our results support that PB projections to the BF and MS specifically regulating cortical and HPC activity.


Subject(s)
Basal Forebrain , Parabrachial Nucleus , Rats , Animals , Wakefulness/physiology , Basal Forebrain/physiology , Arousal/physiology , Electroencephalography , Hippocampus
6.
Sleep Med ; 115: 162-173, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38367358

ABSTRACT

The hippocampus (HPC) plays a pivotal role in fear learning and memory. Our two recent studies suggest that rapid eye movement (REM) sleep via the HPC downregulates fear memory consolidation and promotes fear extinction. However, it is not clear whether and how the dorsal and the ventral HPC regulates fear memory differently; and how the HPC in wake regulates fear memory. By chemogenetic stimulating in the HPC directly and its afferent entorhinal cortex that selectively activated the HPC in REM sleep for 3-6 h post-fear-acquisition, we found that HPC activation in REM sleep consolidated fear extinction memory. In particular, dorsal HPC (dHPC) stimulation in REM sleep virtually eliminated fear memory by enhancing fear extinction and reducing fear memory consolidation. By contrast, chemogenetic stimulating HPC afferent the supramammillary nucleus (SUM) induced 3-hr wake with HPC activation impaired fear extinction. Finally, desipramine (DMI) injection that selectively eliminated REM sleep for >6 h impaired fear extinction. Our results demonstrate that the HPC is critical for fear memory regulation; and wake HPC and REM sleep HPC have an opposite role in fear extinction of respective impairment and consolidation.


Subject(s)
Fear , Memory Consolidation , Humans , Extinction, Psychological/physiology , Sleep/physiology , Learning/physiology , Hippocampus , Memory Consolidation/physiology
7.
Mol Med Rep ; 28(6)2023 Dec.
Article in English | MEDLINE | ID: mdl-37921064

ABSTRACT

Subacute progressive neuropsychiatric symptoms with cognitive and motor impairment and autoimmune seizures are some of the typical symptoms of anti­N­methyl­D­aspartate receptor (anti­NMDAR) encephalitis. The mechanisms underlying this disease are yet to be elucidated, which could be partly attributed to the lack of appropriate animal models. The present study aimed to establish an active immune mouse model of anti­NMDAR encephalitis. Mice were immunized with the extracellular segment of the NMDA1 protein, then subjected to open­field and novel object recognition experiments. Plasma was collected after euthanasia on day 30 after immunization and anti­NMDA1 antibodies were detected using ELISA. Furthermore, brain slices were analyzed to measure postsynaptic density protein 95 (PSD­95) and NMDA1 expression. Western blot analysis of NMDA1 and PSD­95 protein expression levels in the hippocampus was also performed. In addition, protein expression levels of PSD­95 and NMDA1 in mouse neuronal HT­22 cells were evaluated. Compared with controls, mice immunized with NMDA1 exhibited anxiety, depression and memory impairment. Moreover, high anti­NMDA1 antibody titers were detected with ELISA and the levels of anti­NMDA1 antibody reduced postsynaptic NMDA1 protein density in the mouse hippocampus. These findings demonstrated the successful construction of a novel mouse model of anti­NMDAR encephalitis by actively immunizing the mice with the extracellular segment of the NMDA1 protein. This model may be useful for studying the pathogenesis and drug treatment of anti­NMDAR encephalitis in the future.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis , Mice , Animals , Vaccination , Receptors, N-Methyl-D-Aspartate , Disks Large Homolog 4 Protein , Apolipoproteins E
8.
Front Neurosci ; 17: 1122803, 2023.
Article in English | MEDLINE | ID: mdl-36998723

ABSTRACT

Introduction: Fear and sleep impairments common co-exist, but the underlying mechanisms remain unclear. Hypothalamic orexinergic neurons are involved in the regulation of sleep-wake and fear expression. The ventrolateral preoptic area (VLPO) is an essential brain region to promote sleep, and orexinergic axonal fibers projecting to the VLPO are involved in the maintenance of sleep-wake. Neural pathways from hypothalamic orexin neurons to the VLPO might mediate sleep impairments induced by conditioned fear. Methods: To verify above hypothesis, electroencephalogram (EEG) and electromyogram (EMG) were recorded for analysis of sleep-wake states before and 24 h after conditioned fear training. The retrograde tracing technique and immunofluorescence staining was used to identify the projections from the hypothalamic orexin neurons to the VLPO and to observe their activation in mice with conditioned fear. Moreover, optogenetic activation or inhibition of hypothalamic orexin-VLPO pathways was performed to observe whether the sleep-wake can be regulated in mice with conditioned fear. Finally, orexin-A and orexin receptor antagonist was administered into the VLPO to certify the function of hypothalamic orexin-VLPO pathways on mediating sleep impairments induced by conditioned fear. Results: It was found that there was a significant decrease in the non-rapid eye movement (NREM) and rapid eye movement (REM) sleep time and a significant increase in the wakefulness time in mice with conditioned fear. The results of retrograde tracing technique and immunofluorescence staining showed that hypothalamic orexin neurons projected to the VLPO and observed the CTB labeled orexin neurons were significantly activated (c-Fos+) in the hypothalamus in mice with conditioned fear. Optogenetic activation of hypothalamic orexin to the VLPO neural pathways significantly decreased NREM and REM sleep time and increased wakefulness time in mice with conditioned fear. A significant decrease in NREM and REM sleep time and an increase in wakefulness time were observed after the injection of orexin-A into the VLPO, and the effects of orexin-A in the VLPO were blocked by a pre-administrated dual orexin antagonist (DORA). Conclusion: These findings suggest that the neural pathways from hypothalamic orexinergic neurons to the VLPO mediate sleep impairments induced by conditioned fear.

9.
Int J Biol Macromol ; 233: 123652, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36780962

ABSTRACT

Neuropsychiatric diseases are related to early life stress (ELS), patients often have abnormal learning, memory and emotion. But the regulatory mechanism is unclear. Hippocampal synaptic plasticity (HSP) changes are important mechanism. RhoA pathway is known to regulate HSP by modulating of dendritic spines (DS), whether it's involved in HSP changes in ELS hasn't been reported. So we investigated whether and how RhoA participates in HSP regulation in ELS. The ELS model was established by separation-rearing in juvenile. Results of IntelliCage detection etc. showed simple learning and memory wasn't affected, but spatial, punitive learning and memories reduced, the desire to explore novel things reduced, the anxiety-like emotion increased. We further found hippocampus was activated, the hippocampal neurons dendritic complexities reduced, the proportion of mature DS decreased. The full-length transcriptome sequencing techniques was used to screen for differentially expressed genes involved in regulating HSP changes, we found RhoA gene was up-regulated. We detected RhoA protein, RhoA phosphorylation and downstream molecules expression changes, results shown RhoA and p-RhoA, p-ROCK2 expression increased, p-LIMK, p-cofilin expression and F-actin/G-actin ratio decreased. Our study revealed HSP changes in ELS maybe regulate by activation RhoA through ROCK2/LIMK/cofilin pathway regulated F-actin/G-actin balance and DS plasticity, affecting emotion and cognition.


Subject(s)
Actins , rhoA GTP-Binding Protein , Animals , Rats , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Cognition , Emotions , Hippocampus/metabolism , Neuronal Plasticity , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
10.
Sci Rep ; 12(1): 20349, 2022 11 27.
Article in English | MEDLINE | ID: mdl-36437277

ABSTRACT

Currently, multiturn dialogue models generate human-like responses based on pretrained language models given a dialogue history. However, most existing models simply concatenate dialogue histories, which makes it difficult to maintain a high degree of consistency throughout the generated text. We speculate that this is because the encoder ignores information about the hierarchical structure between sentences. In this paper, we propose a novel multiturn dialogue generation model that captures contextual information at the sentence level and at the discourse level during the encoding process. The context semantic information is dynamically modeled through a difference-aware module. A sentence order prediction training task is also designed to learn representation by reconstructing the order of disrupted sentences with a learning-to-rank algorithm. Experiments on the multiturn dialogue dataset, DailyDialog, demonstrate that our model substantially outperforms the baseline model in terms of both automatic and human evaluation metrics, generating more fluent and informative responses than the baseline model.


Subject(s)
Language , Semantics , Humans , Learning , Algorithms
11.
Oxid Med Cell Longev ; 2022: 9253916, 2022.
Article in English | MEDLINE | ID: mdl-35571236

ABSTRACT

Spinal cord injury (SCI) is a progressive neurodegenerative disease in addition to a traumatic event. Cognitive dysfunction following SCI has been widely reported in patients and animal models. However, the neuroanatomical changes affecting cognitive function after SCI, as well as the mechanisms behind these changes, have so far remained elusive. Herein, we found that SCI accelerates oxidative stress damage of hippocampal neuronal mitochondria. Then, for the first time, we presented a three-dimensional morphological atlas of rat hippocampal neurons generated using a fluorescence Micro-Optical Sectioning Tomography system, a method that accurately identifies the spatial localization of neurons and trace neurites. We showed that the number of dendritic branches and dendritic length was decreased in late stage of SCI. Western blot and transmission electron microscopy analyses also showed a decrease in synaptic communication. In addition, a battery of behavioral tests in these animals revealed hippocampal based cognitive dysfunction, which could be attributed to changes in the dendritic complexity of hippocampal neurons. Taken together, these results suggested that mitochondrial abnormalities in hippocampal neurons induced the dendritic complexity reduction and cognitive decline following SCI. Our study highlights the neuroanatomical basis and importance of mitochondria in brain degeneration following SCI, which might contribute to propose new therapeutic strategies.


Subject(s)
Cognitive Dysfunction , Neurodegenerative Diseases , Spinal Cord Injuries , Animals , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Humans , Mitochondria , Neurodegenerative Diseases/metabolism , Rats , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
12.
PeerJ ; 10: e13605, 2022.
Article in English | MEDLINE | ID: mdl-35769142

ABSTRACT

Background: Numerous studies have shown that exposure to prenatal maternal stress (PMS) is associated with various psychopathological outcomes of offspring. The accumulating evidence linking bacteria in the gut and neurons in the brain (the microbiota-gut-brain axis) has been aconsensus; however, there is a lack of research on the involvement mechanism of gut microbiota in the regulation of the BDNF/CREB signaling pathway in the hippocampus of prenatally stressed offspring. Methods: Pregnant rats were subjected to chronic unpredictable mild stress (CUMS) to establish the prenatal maternal stress model. The body weight was measured and the behavioral changes were recorded. Offspring were tested to determine emotional state using sucrose preference test (SPT), open-field test (OFT) and suspended tail test (STT). Gut microbiota was evaluated by sequencing the microbial 16S rRNA V3-V4 region, and the interactive analysis of bacterial community structure and diversity was carried out. The expression of hippocampal BDNF, TrkB and CREB mRNA and proteins were respectively measured using RT-PCR and Western blotting. Results: Prenatal maternal stress increased maternal plasma corticosterone levels, slowed maternal weight gain and caused depression-like behaviors (all P < 0.05). In offspring, prenatal maternal stress increased plasma corticosterone levels (P < 0.05) and emotional behavior changes (depression-like state) were observed (P < 0.05). The species abundance, diversity and composition of the offspring's gut microbiota changed after the maternal stress during pregnancy (P < 0.05). Compared with the control group's offspring, the species abundance of Lactobacillaceae was dropped, while the abundance of the Muribaculaceae species abundance was risen. Concurrent, changes in the hippocampal structure of the offspring and decreases in expression of BDNF/CREB signaling were noted (P < 0.05). Conclusions: Prenatal maternal stress leads to high corticosterone status and abnormal emotion behavior of offspring, which may be associated with the abnormal BDNF/CREB signaling in hippocampus of offspring caused by the change of gut microbiota composition.


Subject(s)
Brain-Derived Neurotrophic Factor , Gastrointestinal Microbiome , Animals , Female , Pregnancy , Rats , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone , Emotions , Hippocampus/metabolism , RNA, Ribosomal, 16S/metabolism , Signal Transduction
13.
Sci Rep ; 11(1): 22750, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815423

ABSTRACT

Generating fluent, coherent, and informative text from structured data is called table-to-text generation. Copying words from the table is a common method to solve the "out-of-vocabulary" problem, but it's difficult to achieve accurate copying. In order to overcome this problem, we invent an auto-regressive framework based on the transformer that combines a copying mechanism and language modeling to generate target texts. Firstly, to make the model better learn the semantic relevance between table and text, we apply a word transformation method, which incorporates the field and position information into the target text to acquire the position of where to copy. Then we propose two auxiliary learning objectives, namely table-text constraint loss and copy loss. Table-text constraint loss is used to effectively model table inputs, whereas copy loss is exploited to precisely copy word fragments from a table. Furthermore, we improve the text search strategy to reduce the probability of generating incoherent and repetitive sentences. The model is verified by experiments on two datasets and better results are obtained than the baseline model. On WIKIBIO, the result is improved from 45.47 to 46.87 on BLEU and from 41.54 to 42.28 on ROUGE. On ROTOWIRE, the result is increased by 4.29% on CO metric, and 1.93 points higher on BLEU.

14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 37(1): 24-30, 2021 Jan.
Article in Zh | MEDLINE | ID: mdl-33441225

ABSTRACT

Objective To study the therapeutic effect of rapamycin (RAPA) on experimental autoimmune myasthenia gravis (EAMG) rats and to explore the related immune mechanisms. Methods The mouse-derived acetylcholine receptor alpha subunit 97-116 peptide (R97-116) was used to immunize Lewis rats to establish an EAMG rat model. The rats were randomly divided into three groups: complete Freund's adjuvant control group (CFA group), EAMG model control group, and RAPA treatment group [1 mg/(kg.d)]. The Lennon muscle strength scoring scale was used to evaluate rats' clinical symptoms in each group once every two days, and their body mass was recorded. ELISA was performed to detect the level of anti-R97-116 antibodies in the peripheral blood of rats. Flow cytometry was used to detect the numbers of Th17 cells and regulatory T cells (Tregs) in rat splenocytes. Splenocytes were stimulated with 5 µg/mL concanavalin A (ConA), 10 µg/mL R97-116 and RPMI1640 medium, and the proliferation activity of rat splenocytes was tested by CCK-8 assay. Results RAPA treatment significantly improved the body mass and clinical scores in EAMG rats. Compared with the CFA group, the number of Th17 cells in the spleen of the EAMG group increased, and the number of Tregs decreased. Compared with the EAMG group, the number of Th17 cells in the spleen of RAPA-treated rats significantly dropped, the number of Tregs went up, and the level of anti-R97-116 antibodies in the serum went down. RAPA treatment inhibited the proliferation of lymphocytes induced by RPMI1640 medium, R97-116, and ConA stimulation. Conclusion RAPA may alleviate the clinical symptoms of EAMG rats by down-regulating the ratio of Th17 cells/Tregs.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental , T-Lymphocytes, Regulatory , Animals , Mice , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Rats , Rats, Inbred Lew , Sirolimus/pharmacology , Th17 Cells
15.
Toxicon ; 194: 70-78, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33610632

ABSTRACT

Cone snails are predatory gastropod mollusks that are distributed in all tropical marine environments and contain small peptides (conotoxins) in their venom to capture prey. However, the biochemical and molecular aspects of conotoxins remain poorly understood. In this article, a novel α4/7-conotoxin, Lv1d, was obtained from the venom duct cDNA library of the worm-hunting Conus lividus collected from the South China Sea. The cDNA of Lv1c encodes a 65 residue conopeptide precursor, which consists of a 21 residue signal peptide, a 27 residue Pro region, and 17 residues of mature peptide. The mature peptide Lv1d was chemically synthesized according to the sequence GCCSDPPCRHKHQDLCG. It was found that 10 µM Lv1d can completely inhibit frog sciatic nerve-gastrocnemius muscle contractility within 60 min. Moreover, 100 µg/kg Lv1d showed good analgesic effects in mouse hot plate model and formalin test. Patch clamp experiments showed that 5 µM Lv1d can inhibit the cholinergic microexcitatory postsynaptic currents (mEPSCs) requency and amplitude of projection neurons in Drosophila. In conclusion, the synthesis of Lv1d and its biological and physiological data might contribute to the development of this peptide as a novel potential drug for therapeutic applications. This finding also expands the knowledge of the targeting mechanism of the α4/7-subfamily conotoxins.


Subject(s)
Conotoxins , Conus Snail , Amino Acid Sequence , Analgesics/pharmacology , Animals , China , Conotoxins/pharmacology , Mice
16.
Neurosci Lett ; 728: 134978, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32302699

ABSTRACT

The steroid hormone 17ß-estradiol (estrogen) exerts neuroprotective effects in several types of neurological disorders including epilepsy. The novel G protein-coupled estrogen receptor 1 (GPER1), also called GPR30, mediates the non-genomic effects of 17ß-estradiol. However, the specific role of GPER1 in status epilepticus (SE) remains unclear. In this report, we evaluated the effects of GPER1 on the hippocampus during SE and the underlying mechanism was studied. Our results revealed that pilocarpine-induced GPER1-KD epileptic rats exhibited a shorter latency to generalized convulsions and strikingly elevated seizure severity. Additionally, the electroencephalographic seizure activity also corresponded to these results. Fast-Fourier analysis indicated an enhancement of power in the theta and alpha bands during SE in GPER1-KD rats. In addition, epilepsy-induced pathological changes were dramatically exacerbated in GPER1-KD rats, including neuron damage and neuroinflammation in hippocampus. GPER1 might be associated with the susceptibility to and severity of epileptic seizures. In summary, our results suggested that GPER1 plays a neuroprotective role in SE, and might be a candidate target for epilepsy therapy.


Subject(s)
Hippocampus/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Status Epilepticus/metabolism , Animals , Electroencephalography , Estradiol/pharmacology , Estrogens/metabolism , Estrogens/pharmacology , Male , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy
17.
Drug Deliv ; 24(1): 651-659, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28283003

ABSTRACT

Nano-scaled materials have been proved to be ideal DNA carriers for transgene. Bacterial magnetic particles (BMPs) help to reduce the toxicity of polyethylenimine (PEI), an efficient gene-transferring agent, and assist tissue transgene ex vivo. Here, the effectiveness of the BMP-PEI complex-conjugated foreign DNAs (BPDs) in promoting testes-mediated gene transfer (TMGT) in mouse was compared with that of liposome-conjugated foreign DNAs. The results proved that through testes injection, the clusters of BPDs successfully reached the cytoplasm and the nuclear of spermatogenesis cell, and expressed in testes of transgene founder mice. Additionally, the ratio of founder mice obtained from BPDs (88%) is about 3 times higher than the control (25%) (p < 0.05). Interestingly, the motility of sperms recovered from epididymis of the founder mice from BPD group were significantly improved, as compared with the control (p < 0.01). Based on classic breeding, the ratio of transgene mice within the first filial was significantly higher in BPDs compared with the control (73.8% versus 11.6%, p < 0.05). TMGT in this study did not produce visible histological changes in the testis. In conclusion, nano-scaled BPDs could be an alternative strategy for efficiently producing transgene mice in vivo.


Subject(s)
Gene Transfer Techniques , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Magnetosomes/genetics , Magnetospirillum/genetics , Spermatozoa/metabolism , Testis/metabolism , Transgenes , Animals , Founder Effect , Gene Expression Regulation , Genotype , Imines/chemistry , Imines/metabolism , Liposomes , Magnetosomes/metabolism , Magnetospirillum/metabolism , Male , Mice , Mice, Transgenic , Phenotype , Polyethylenes/chemistry , Polyethylenes/metabolism , Sperm Motility , Spermatogenesis , Testis/cytology , Time Factors
18.
Sci Rep ; 6: 33428, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27641997

ABSTRACT

This study examined sustained co-delivery of vascular endothelial growth factor (VEGF), angiopoietin-1 and basic fibroblast growth factor (bFGF) encapsulated in angiogenic microspheres. These spheres were delivered to sites of spinal cord contusion injury in rats, and their ability to induce vessel formation, neural regeneration and improve hindlimb motor function was assessed. At 2-8 weeks after spinal cord injury, ELISA-determined levels of VEGF, angiopoietin-1, and bFGF were significantly higher in spinal cord tissues in rats that received angiogenic microspheres than in those that received empty microspheres. Sites of injury in animals that received angiogenic microspheres also contained greater numbers of isolectin B4-binding vessels and cells positive for nestin or ß III-tubulin (P < 0.01), significantly more NF-positive and serotonergic fibers, and more MBP-positive mature oligodendrocytes. Animals receiving angiogenic microspheres also suffered significantly less loss of white matter volume. At 10 weeks after injury, open field tests showed that animals that received angiogenic microspheres scored significantly higher on the Basso-Beattie-Bresnahan scale than control animals (P < 0.01). Our results suggest that biodegradable, biocompatible PLGA microspheres can release angiogenic factors in a sustained fashion into sites of spinal cord injury and markedly stimulate angiogenesis and neurogenesis, accelerating recovery of neurologic function.


Subject(s)
Microspheres , Motor Activity/physiology , Neovascularization, Physiologic , Nerve Regeneration/physiology , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Animals , Anisotropy , Axons/metabolism , Axons/ultrastructure , Ephrin-A3/metabolism , Female , Lactic Acid/chemistry , Magnetic Resonance Imaging , MicroRNAs/genetics , MicroRNAs/metabolism , Neural Stem Cells/metabolism , Organ Size , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Up-Regulation/genetics , White Matter/pathology , White Matter/physiopathology
19.
Int J Dev Neurosci ; 47(Pt B): 333-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26380950

ABSTRACT

Recent studies have shown that GAP-43 is highly expressed in horizontally dividing neural progenitor cells, and G protein complex are required for proper mitotic-spindle orientation of those progenitors in the mammalian developing cortex. In order to verify the hypothesis that GAP-43 may influence the orientation of cell division through interacting with G proteins during neurogenesis, the GAP-43 RNA from adult C57 mouse was cloned into the pEGFP-N1 vector, which was then transfected into Madin-Darby Canine Kidney (MDCK) cells cultured in a three-dimensional (3D) cell culture system. The interaction of GAP-43 with Gαi was detected by co-immunoprecipitation (co-IP), while cystogenesis of 3D morphogenesis of MDCK cells and expression of GAP-43 and Gαi were determined by immunofluorescence and Western blotting. The results showed are as follows: After being transfected by pEGFP-N1-GAP-43, GAP-43 was localized on the cell membrane and co-localized with Gαi, and this dramatically induced a defective cystogenesis in 3D morphogenesis of MDCK cells. The functional interaction between GAP-43 and Gαi proteins was proven by the co-IP assay. It can be considered from the results that the GAP-43 is involved in the orientation of cell division by interacting with Gαi and this should be an important mechanism for neurogenesis in the mammalian brain.


Subject(s)
Cell Division/physiology , Cell Polarity/physiology , GAP-43 Protein/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Animals , Cell Culture Techniques , Cell Division/drug effects , Cell Polarity/genetics , Dogs , GAP-43 Protein/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Imaging, Three-Dimensional , Immunoprecipitation , Madin Darby Canine Kidney Cells , Mice , Transfection
20.
Article in Zh | MEDLINE | ID: mdl-14694642

ABSTRACT

OBJECTIVE: To find an inhibitor to reduce the volatilization of formalin. METHOD: The saturated solution of sodium hydrosulphite (SHS) was sprayed on the surface of the anatomy specimens, then the concentration of formaldehyde in the air was tested. RESULTS: The concentration of formaldehyde in the air of SHS sprayed group [(3.10 +/- 1.22) mg/m3] was significantly lower than that of the control group [(8.36 +/- 4.11) mg/m3, P < 0.01]. CONCLUSION: SHS may be a volatilization inhibitor for formalin, which could reduce the concentration of formaldehyde in the air.


Subject(s)
Air Pollution, Indoor/prevention & control , Formaldehyde/analysis , Anatomy , Formaldehyde/chemistry , Sulfites/chemistry , Volatilization
SELECTION OF CITATIONS
SEARCH DETAIL