Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Exp Ther Med ; 25(4): 180, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37006883

ABSTRACT

MicroRNAs (miRNAs or miRs) are commonly involved in regulating myocardial ischemia/reperfusion (I/R) injury by binding and silencing their target genes. However, whether miRNAs regulate myocardial I/R-induced pyroptosis remains unclear. The present study established an in vivo rat model of myocardial I/R injury and in vitro hypoxia/reoxygenation (H/R) injury model in rat primary cardiomyocytes to investigate the function and the underlying mechanisms of miRNAs on I/R injury-induced pyroptosis. RNA sequencing was utilized to select the candidate miRNAs between normal and I/R group. Reverse transcription-quantitative PCR and western blotting were performed to detect candidate miRNAs (miR-30c-5p, also known as miR-30c) and SRY-related high mobility group-box gene 9 (SOX9) expression, as well as expression of pyroptosis-associated proteins (NF-κB, ASC, caspase-1, NLRP3) in the myocardial I/R model. ELISA was used to measure pyroptosis-associated inflammatory markers IL-18 and IL-1ß. Moreover, the link between miR-30c and SOX9 was predicted using bioinformatics and luciferase reporter assay. In myocardial I/R injured rats, miR-30c was downregulated, while the expression of SOX9 was upregulated. Overexpression of miR-30c inhibited pyroptosis both in vivo and in vitro. Furthermore, miR-30c negatively regulated SOX9 expression by binding its 3'untranslated region. In conclusion, the miR-30c/SOX9 axis decreased myocardial I/R injury by suppressing pyroptosis, which may be a potential therapeutic target.

2.
CNS Neurosci Ther ; 29(1): 378-389, 2023 01.
Article in English | MEDLINE | ID: mdl-36353753

ABSTRACT

AIMS: The claustrum has long been regarded as a vital center for conscious control. Electrical stimulation or damage to the claustrum can result in decreased awareness or loss of consciousness, suggesting that the claustrum may be a target for the action of general anesthetics. This study aimed to determine the role of the claustrum in propofol anesthesia. METHODS: We first applied a fiber photometry calcium signal recording system to record the claustral neuronal activity during the entire process of propofol anesthesia. Chemogenetic activation of claustral neurones was then performed to verify their role in anesthesia. Finally, muscimol (GABAa receptor agonist) and gabazine (GABAa receptor antagonist) were microinjected into the claustrum to determine whether their GABAa receptors were involved in modulating propofol anesthesia. EEG and behavioral indicators, such as anesthetic sensitivity and efficacy, were recorded and analyzed. RESULTS: An evident anesthesia-related change in claustrum neuronal activity was suppressed during propofol-induced unconsciousness and restored following recovery from anesthesia. Chemogenetic activation of claustrum neurons results in attenuated propofol sensitivity, a shorter anesthesia duration, and an EEG shift toward wakefulness. Manipulation of GABAa receptors in the claustrum showed bidirectional control of propofol sensitivity, as activation decreases anesthesia efficiency while inactivation augments it. Additionally, inhibiting claustrum GABAa receptors increases cortical EEG slow waves. CONCLUSIONS: Claustrum neurones and their GABAa receptors are implicated in the modulation of propofol anesthesia in both behavioral and EEG assessments. Our findings create scope to reveal the brain targets of anesthetic action further and add to the existing evidence on the consciousness-modulating role of the claustrum.


Subject(s)
Anesthesia , Anesthetics, General , Claustrum , Propofol , Propofol/pharmacology , Receptors, GABA-A , Anesthetics, General/pharmacology , Electroencephalography
3.
iScience ; 25(9): 104989, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36093041

ABSTRACT

Neonatal repetitive noxious stimuli (RNS) has been shown to cause long-term harmful effects on nociceptive processing, learning, and memory which persist until adulthood. Plasticity-related gene 1 (PRG-1) regulates synaptic plasticity and functional reorganization in the brain during neuronal development. In this study, neonatal RNS rats were established by repetitive needle pricks to neonatal rats on all four feet to model repetitive pain exposure in infants. Neonatal RNS caused thermal hyperalgesia, mechanical allodynia, learning, and memory impairments which manifested in young rats and persisted until adulthood. Hippocampal PRG-1/N-ethylmaleimide sensitive fusion protein (NSF) interaction was determined to be responsible for the RNS-induced impairment via enhanced extracellular glutamate release and AMPAR GluR2 trafficking deficiency in a cell-autonomous manner. These pathways likely act synergistically to cause changes in dendritic spine density. Our findings suggest that PRG-1 prevents the RNS-induced hyperalgesia, learning, and memory impairment by regulating synaptic plasticity via NSF/Glu/GluR2 signaling.

4.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35022234

ABSTRACT

Amino acids are essential for cell growth and metabolism. Amino acid and growth factor signaling pathways coordinately regulate the mechanistic target of rapamycin complex 1 (mTORC1) kinase in cell growth and organ development. While major components of amino acid signaling mechanisms have been identified, their biological functions in organ development are unclear. We aimed to understand the functions of the critically positioned amino acid signaling complex GAP activity towards Rags 2 (GATOR2) in brain development. GATOR2 mediates amino acid signaling to mTORC1 by directly linking the amino acid sensors for arginine and leucine to downstream signaling complexes. Now, we report a role of GATOR2 in oligodendrocyte myelination in postnatal brain development. We show that the disruption of GATOR2 complex by genetic deletion of meiosis regulator for oocyte development (Mios, encoding a component of GATOR2) selectively impairs the formation of myelinating oligodendrocytes, thus brain myelination, without apparent effects on the formation of neurons and astrocytes. The loss of Mios impairs cell cycle progression of oligodendrocyte precursor cells, leading to their reduced proliferation and differentiation. Mios deletion manifests a cell type-dependent effect on mTORC1 in the brain, with oligodendroglial mTORC1 selectively affected. However, the role of Mios/GATOR2 in oligodendrocyte formation and myelination involves mTORC1-independent function. This study suggests that GATOR2 coordinates amino acid and growth factor signaling to regulate oligodendrocyte myelination.


Subject(s)
Amino Acids/metabolism , Brain/metabolism , Multiprotein Complexes/metabolism , Myelin Sheath/metabolism , Signal Transduction , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Gene Deletion , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout , Models, Biological , Neural Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Transgenes
5.
Int J Neurosci ; 132(8): 802-814, 2022 Aug.
Article in English | MEDLINE | ID: mdl-33174773

ABSTRACT

PURPOSE: The aim of the current study was to explore the role of the basal forebrain (BF) in propofol anaesthesia. METHODS: In the present study, we observed the neural activities of the BF during propofol anaesthesia using calcium fibre photometry recording. Subsequently, ibotenic acid was injected into the BF to verify the role of the BF in propofol anaesthesia. Finally, to test whether GABAA receptors in the BF were involved in modulating propofol anaesthesia, muscimol (GABAA receptor agonist) and gabazine (GABAA receptor antagonist) were microinjected into the BF. Cortical electroencephalogram (EEG), time to loss of righting reflex (LORR), and recovery of righting reflex (RORR) under propofol anaesthesia were recorded and analysed. RESULTS: The activity of BF neurons was inhibited during induction of propofol anaesthesia and activated during emergence from propofol anaesthesia. In addition, non-specifical lesion of BF neurons significantly prolonged the time to RORR and increased delta power in the frontal cortex under propofol anaesthesia. Next, microinjection of muscimol into the BF delayed emergence from propofol anaesthesia, increased delta power of the frontal cortex, and decreased gamma power under propofol anaesthesia. Conversely, infusion of gabazine accelerated emergence times and decreased EEG delta power. CONCLUSIONS: The basal forebrain is involved in modulating frontal cortex delta activity and emergence from propofol anaesthesia. Additionally, the GABAA receptors in the basal forebrain are involved in regulating emergence propofol anaesthesia.


Subject(s)
Anesthesia , Basal Forebrain , Propofol , Animals , Basal Forebrain/metabolism , Electroencephalography , Muscimol/pharmacology , Propofol/pharmacology , Rats , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/pharmacology
6.
CNS Neurosci Ther ; 27(7): 792-804, 2021 07.
Article in English | MEDLINE | ID: mdl-33764684

ABSTRACT

AIMS: The basal forebrain (BF) plays an essential role in wakefulness and cognition. Two subtypes of BF gamma-aminobutyric acid (GABA) neurons, including somatostatin-expressing (GABASOM ) and parvalbumin-positive (GABAParv ) neurons, function differently in mediating the natural sleep-wake cycle. Since the loss of consciousness induced by general anesthesia and the natural sleep-wake cycle probably share similar mechanisms, it is important to clarify the accurate roles of these neurons in general anesthesia procedure. METHODS: Based on two transgenic mouse lines expressing SOM-IRES-Cre and PV-IRES-Cre, we used a combination of genetic activation, inactivation, and chronic ablation approaches to further explore the behavioral and electroencephalography (EEG) roles of BFSOM and BFParv neurons in general anesthesia. After a single intravenous injection of propofol and the induction and recovery times of isoflurane anesthesia, the anesthesia time was compared. The changes in cortical EEG under different conditions were also compared. RESULTS: Activation of BF GABASOM neurons facilitates both the propofol and isoflurane anesthesia, manifesting as a longer anesthesia duration time with propofol anesthesia and a fast induction time and longer recovery time with isoflurane anesthesia. Moreover, BF GABASOM -activated mice displayed a greater suppression of cortical electrical activity during anesthesia, showing an increase in δ power bands or a simultaneous decrease in high-frequency power bands. However, only a limited and nuanced effect on propofol and isoflurane anesthesia was observed with the manipulated BF GABAParv neurons. CONCLUSIONS: Our results suggested that BF GABASOM neurons play a critical role in propofol and isoflurane general anesthesia, while BF GABAParv neurons appeared to have little effect.


Subject(s)
Anesthesia, General/methods , Basal Forebrain/metabolism , GABAergic Neurons/metabolism , Isoflurane/pharmacology , Parvalbumins/metabolism , Propofol/pharmacology , Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Animals , Basal Forebrain/drug effects , Electroencephalography/methods , GABAergic Neurons/drug effects , Male , Mice , Mice, 129 Strain , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism
7.
Cell Cycle ; 20(4): 434-444, 2021 02.
Article in English | MEDLINE | ID: mdl-33522393

ABSTRACT

Tumor-associated macrophages contribute to cell growth, development, and metastasis in various cancers. However, the underlying mechanisms of M2 macrophage that modulate the progression of gastric cancer (GC) remain largely unknown. In this study, we detected the ratio of macrophages in GC tissues and found that the proportion of M2 macrophages was increased in GC tissues. We then co-cultured GC cells with M1 and M2 macrophages, respectively, and then assessed cell proliferation and tumorigenicity of GC cells by MTT and colony formation assay. The results indicated that M2 macrophages promoted the proliferation of GC cells, but M1 not. Besides, GW4869, an exosomes inhibitor, reduced the effects induced by M2 macrophage. Then, we isolated and identified exosomes derived from M1 and M2 macrophage, and confirmed that the exosomes could be taken up by GC cells. We demonstrated that M2 macrophage-exosomes could induce the proliferation and tumorigenesis in vitro and in vivo. Moreover, miR-487a was enriched in M2 macrophage-exosomes and further determined that miR-487a exert the functions by targeting TIA1. In conclusion, exosomal miR-487a derived from M2 macrophage promotes the proliferation and tumorigenesis in gastric cancer, and the novel findings might be helpful to the development of novel diagnostic and therapeutic methods in GC.


Subject(s)
Disease Progression , Exosomes/metabolism , Macrophages/metabolism , MicroRNAs/biosynthesis , Stomach Neoplasms/metabolism , Animals , Cell Proliferation/physiology , Coculture Techniques , Exosomes/genetics , Exosomes/pathology , Humans , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
8.
Acta Biochim Biophys Sin (Shanghai) ; 53(3): 333-341, 2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33522578

ABSTRACT

Metformin is a widely prescribed hypoglycemic drug. Many studies have shown its anti-cancer properties. In the present study, we aimed to explore the effect of metformin on breast cancer and clarify the underlying mechanism. Our results showed that metformin induced ferroptosis in MDA-MB-231 cells through upregulating miR-324-3p expression. Overexpression of miR-324-3p inhibited cancer cell viability. miR-324-3p inhibitor promoted cell viability. Further studies showed that the effect of miR-324-3p was mediated by directly targeting glutathione peroxidase 4 (GPX4). miR-324-3p bound to the 3'-UTR of GPX4 and led to the downregulation of GPX4. In vivo studies showed that metformin induced ferroptosis by upregulating miR-324-3p in the xenograft model of breast cancer in mice. Our study suggested that metformin promotes ferroptosis of breast cancer by targeting the miR-324-3p/GPX4 axis. Metformin could act as a potential anti-cancer agent through the induction of ferroptosis.


Subject(s)
Ferroptosis/drug effects , Metformin/pharmacology , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , RNA, Neoplasm/metabolism , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , Neoplasm Proteins/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , RNA, Neoplasm/genetics
9.
Acta Biochim Biophys Sin (Shanghai) ; 53(3): 325-332, 2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33501502

ABSTRACT

Glioma is one of the most pervasive and invasive primary malignancies in the central nervous system. Due to its abnormal proliferation, glioma remains hard to cure at present. Protein tyrosine phosphatase 1B (PTP1B) has been proved to be involved in the process of proliferation in many malignancies. However, whether PTP1B is involved in the proliferation of glioma and how it acts are still unclear. In this study, the PTP1B expressions in glioma tissues and cells were determined by quantitative real-time PCR and western blot analysis. The effects of PTP1B on the proliferation characteristics of glioma were explored using 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation assay, and tumor xenografts in mice. We found that the protein and mRNA levels of PTP1B in glioma tissues were significantly higher than those in paired nontumor tissues. MTT and clone formation assays showed that PTP1B is closely related to human glioma cell proliferation. In addition, TargetScan revealed that miR-34c regulates PTP1B. Mechanistically, we proved that miR-34c negatively regulates PTP1B and then participates in the regulation of glioma cell proliferation in vivo. Collectively, these results suggested that miR-34c inhibits the proliferation of human glioma cells by targeting PTP1B, which will provide a potential target for the treatment of glioma.


Subject(s)
Cell Proliferation , Glioma/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , RNA, Neoplasm/metabolism , Animals , Cell Line, Tumor , Glioma/genetics , Glioma/pathology , Humans , Mice , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , RNA, Neoplasm/genetics
10.
RNA Biol ; 18(9): 1279-1290, 2021 09.
Article in English | MEDLINE | ID: mdl-33151124

ABSTRACT

Treatment of central nervous system (CNS) demyelination is greatly hindered by lack of the knowledge regarding to underlying molecular mechanisms as well as therapeutic agents. Here, we report a novel small molecule agent, gastrodin (GAS), which can significantly promote CNS myelination in in vivo mice models. By using high-throughput sequencing analysis, we discover a key long non-coding RNA Gm7237 that can enhance CNS myelination and is up-regulated by GAS. Through using bioinformatic analysis and experimental validations, we further unravel that microRNA-142a (miR-142a) and its target myelin gene regulatory factor (MRF) is under the direct regulation by Gm7237. Finally, we demonstrate that Gm7237/miR-142a/MRF axis is the key pathway involved in CNS myelination mediated by GAS. Overall, our results provide not only a novel agent for therapeutic treatment of CNS demyelination but also a molecular basis responsible for GAS-promoted CNS myelination.


Subject(s)
Benzyl Alcohols/pharmacology , Central Nervous System/cytology , Gene Expression Regulation/drug effects , Glucosides/pharmacology , MicroRNAs/genetics , Myelin Sheath/physiology , RNA, Long Noncoding/genetics , Transcription Factors/metabolism , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Mice , Mice, Inbred C57BL , Myelin Sheath/drug effects , Transcription Factors/genetics
11.
Front Neurosci ; 14: 559077, 2020.
Article in English | MEDLINE | ID: mdl-33192246

ABSTRACT

Cholinergic neurons in the basal forebrain (BF) have long been considered to be the key neurons in the regulation of cortical and behavioral arousal, and cholinergic activation in the downstream region of the BF can arouse anesthetized rats. However, whether the activation of BF cholinergic neurons can induce behavior and electroencephalogram (EEG) recovery from anesthesia is unclear. In this study, based on a transgenic mouse line expressing ChAT-IRES-Cre, we applied a fiber photometry system combined with GCaMPs expression in the BF and found that both isoflurane and propofol inhibit the activity of BF cholinergic neurons, which is closely related to the consciousness transition. We further revealed that genetic lesion of BF cholinergic neurons was associated with a markedly increased potency of anesthetics, while designer receptor exclusively activated by designer drugs (DREADD)-activated BF cholinergic neurons was responsible for slower induction and faster recovery of anesthesia. We also documented a significant increase in δ power bands (1-4 Hz) and a decrease in ß (12-25 Hz) power bands in BF cholinergic lesioned mice, while there was a clearly noticeable decline in EEG δ power of activated BF cholinergic neurons. Moreover, sensitivity to anesthetics was reduced after optical stimulation of BF cholinergic cells, yet it failed to restore wake-like behavior in constantly anesthetized mice. Our results indicate a functional role of BF cholinergic neurons in the regulation of general anesthesia. Inhibition of BF cholinergic neurons mediates the formation of unconsciousness induced by general anesthetics, and their activation promotes recovery from the anesthesia state.

12.
Oxid Med Cell Longev ; 2020: 4957238, 2020.
Article in English | MEDLINE | ID: mdl-32963696

ABSTRACT

Nowadays, reperfusion is still the most effective treatment for ischemic heart disease. However, cardiac reperfusion therapy would lead to reperfusion injury, which may have resulted from endoplasmic reticulum stress (ERS) during reperfusion. Diazoxide (DZ) is a highly selective mitochondrial adenosine triphosphate-sensitive potassium channel opener. Its protective effect on I/R injury has been confirmed in many organs such as the heart and brain. However, the mechanism of its protective effect has not been fully elucidated. MicroRNAs (miRNAs) are widely involved in pathologies of heart disease. In this study, we found that miR-10a expression was highly upregulated in the myocardial I/R groups, and DZ treatment significantly reduced the expression of miR-10a. More importantly, we found that DZ treatment can moderate ERS via regulation of the miR-10a/IRE1 pathway in the I/R and H/R models, thereby protecting myocardial H/R injury.


Subject(s)
Cardiotonic Agents/therapeutic use , Diazoxide/therapeutic use , Endoplasmic Reticulum Stress , Membrane Proteins/metabolism , MicroRNAs/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Cardiotonic Agents/pharmacology , Diazoxide/pharmacology , Endoplasmic Reticulum Stress/drug effects , Female , Gene Expression Regulation/drug effects , Male , MicroRNAs/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Up-Regulation/drug effects
13.
Front Neurosci ; 14: 364, 2020.
Article in English | MEDLINE | ID: mdl-32410945

ABSTRACT

Electrical synapses between neurons exhibit a high degree of plasticity, which makes critical contributions to neuronal communication. The GABAergic parvalbumin-expressing (PV+) neurons in the thalamic reticular nucleus (TRN) interact with each other through electrical and chemical synapses. Plasticity of electrical synaptic transmission in TRN plays a key role in regulating thalamocortical and corticothalamic circuits and even the formation of consciousness. We here examined the effects of propofol, a commonly used general anesthetic agent, on the strength of electrical synapses between TRN PV+ neurons by fluorescence-guided patch-clamp recording and pharmacological methods. Results show that 100 µM propofol reduced the electrical synaptic strength between TRN PV+ neurons. Notably, the propofol-induced depression of electrical synaptic strength between TRN PV+ neurons was diminished by saclofen (10 µM, antagonist of GABAB receptors), but not blocked by gabazine (10 µM, antagonist of GABAA receptors). Application of baclofen (10 µM, agonist of GABAB receptors), similar to propofol, also reduced the electrical synaptic strength between TRN PV+ neurons. Moreover, the propofol-induced depression of electrical synaptic strength between TRN PV+ neurons was abolished by 9-CPA (100 µM, specific adenylyl cyclase inhibitor), and by KT5720 (1 µM, selective inhibitor of PKA). Our findings indicate that propofol acts on metabotropic GABAB receptors, resulting in a depression of electrical synaptic transmission of coupled TRN PV+ neurons, which is mediated by the adenylyl cyclase-cAMP-PKA signaling pathway. Our findings also imply that propofol may change the thalamocortical communication via inducing depression of electrical synaptic strength in the TRN.

14.
J Pain Res ; 12: 2905-2913, 2019.
Article in English | MEDLINE | ID: mdl-31695480

ABSTRACT

Mechanisms of postherpetic neuralgia (PHN) are still not clear. Transcripts such as microRNA (miRNA) and circular RNA (circRNA) in the affected skin may take part in the initiation and development of this neuropathic pain; however, their expression profiles in skins of PHN patients have not been reported. The PHN affected skin and the mirror skin were collected and subjected to miRNA and circRNA microarray, and expression profiles were comparatively analyzed. There were 317 differently expressed miRNAs in PHN affected skin compared with mirror skin (fold change ≥2.0), and 13 of them showed fold change >10 in the PHN skin. Only one circRNA, hsa_circRNA_405463 showed fold change >2 in PHN skin, however, 31 circRNAs with fold change ≥1.5. To evaluate functions of differential miRNAs, their target mRNAs were predicted and bioinformatics analyses including gene ontology, Kyoto Encyclopedia of Genes and Genomes pathway were conducted. Target mRNAs significantly (P<0.05) enriched in 85 pathways, such as FoxO, AMPK, MAPK and pathway. These data reported for the first time that miRNA and circRNA differentially expressed in the PHN skin and these transcripts with abnormal expression could be potential targets to treat PHN.

15.
Front Cell Neurosci ; 13: 240, 2019.
Article in English | MEDLINE | ID: mdl-31244607

ABSTRACT

BACKGROUND: Astrocytes are now considered as crucial modulators of neuronal synaptic transmission. General anesthetics have been found to inhibit astrocytic activities, but it is not clear whether general anesthetics within the clinical concentration range affects the astrocyte-mediated synaptic regulation. METHODS: The effects of propofol, dexmedetomidine, and ketamine within clinically effective ranges on the slow inward currents (SICs) were tested by using the whole-cell recording in acute prefrontal cortex (PFC) slice preparations of rats. Astrocytes culture and HPLC were used to measure the effects of different anesthetics on the glutamate release of astrocytes. RESULTS: Propofol and dexmedetomidine showed no significant effect on the amplitude or frequency of SICs. Ketamine was found to inhibit the frequency of SICs in a concentration-dependent manner. The SICs synchronization rate of paired neurons was inhibited by 30 µM ketamine (from 42.5 ± 1.4% to 9.6 ± 0.8%) and was abolished by 300 µM ketamine. The astrocytic glutamate release induced by DHPG, an agonist of astrocytic type I metabotropic glutamate receptors, was not affected by ketamine, and ifenprodil, a selective antagonist of GluN1/GluN2B receptor, blocked all SICs and enhanced the inhibitory effect of 30 µM ketamine on the frequency of SICs. Ketamine at low concentration (3 µM) could inhibit the frequency of SICs, not the miniature excitatory postsynaptic currents (mEPSCs), and the inhibition rate of SICs was significantly higher than mEPSCs with 30 µM ketamine (44.5 ± 3% inhibition vs. 28.3 ± 6% inhibition). CONCLUSION: Our data indicated that ketamine, not propofol and dexmedetomidine, within clinical concentration range inhibits glutamatergic transmission from astrocytes to neurons, which is likely mediated by the extrasynaptic GluN1/GluN2B receptor activation.

16.
FASEB J ; 33(6): 7252-7260, 2019 06.
Article in English | MEDLINE | ID: mdl-30860868

ABSTRACT

Electroencephalogram monitoring during propofol (PRO) anesthesia typically features low-frequency oscillations, which may be involved with thalamic reticular nucleus (TRN) modulation. TRN receives noradrenergic inputs from the locus coeruleus (LC). We hypothesized that specific noradrenergic connections in the TRN may contribute to the emergence from PRO anesthesia. Intranuclei norepinephrine (NE) injections (n = 10) and designer receptors exclusively activated by designer drugs (DREADDs) (n = 10) were used to investigate the role of noradrenergic inputs from the LC to the TRN during PRO anesthesia. Whole-cell recording in acute brain slice preparations was used to identify the type of adrenoceptor that regulates noradrenergic innervation in the TRN. An intracerebral injection of NE into the TRN delays arousal in mice recovering from PRO anesthesia (means ± sd; 486.6 ± 57.32 s for the NE injection group vs. 422.4 ± 48.19 s for the control group; P = 0.0143) and increases the cortical-δ (0.1-4 Hz, 25.4 ± 2.9 for the NE injection group vs. 21.0 ± 1.7 for the control group; P = 0.0094) oscillation. An intra-TRN injection of NE also decreased the EC50 of PRO-induced unconsciousness (57.05 ± 1.78 mg/kg for the NE injection group vs. 72.44 ± 3.23 mg/kg for the control group; P = 0.0096). Moreover, the activation of LC-noradrenergic nerve terminals in the TRN using DREADDs increased the recovery time [466.1 ± 44.57 s for the clozapine N-oxide (CNO) injection group vs. 426.1 ± 38.75 s for the control group; P = 0.0033], decreased the EC50 of PRO-induced unconsciousness (64.77 ± 3.40 mg/kg for the CNO injection group vs. 74.00 ± 2.08 mg/kg for the control group; P = 0.0081), and increased the cortical-δ oscillation during PRO anesthesia (23.29 ± 2.58 for the CNO injection group vs. 19.56 ± 1.9 for the control group; P = 0.0213). In addition, whole-cell recording revealed that NE augmented the inhibitory postsynaptic currents in the TRN neurons via the α1-adrenoceptor. Our data indicated that enhanced NE signaling at the noradrenergic terminals of the LC-TRN projection delays arousal from general anesthesia, which is likely mediated by the α1-adrenoceptor activation. Our findings open a door for further understanding of the functions of various LC targets in both anesthesia and arousal.-Zhang, Y., Fu, B., Liu, C., Yu, S., Luo, T., Zhang, L., Zhou, W., Yu, T. Activation of noradrenergic terminals in the reticular thalamus delays arousal from propofol anesthesia in mice.


Subject(s)
Adrenergic Neurons/physiology , Anesthesia, General , Arousal/physiology , Delayed Emergence from Anesthesia/physiopathology , Intralaminar Thalamic Nuclei/physiopathology , Nerve Endings/physiology , Receptors, Adrenergic, alpha-1/physiology , Adrenergic Neurons/drug effects , Anesthetics, Intravenous , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Designer Drugs/pharmacology , Electroencephalography , Genetic Vectors/administration & dosage , Intralaminar Thalamic Nuclei/drug effects , Mice , Mice, Inbred C57BL , Nerve Endings/drug effects , Norepinephrine/pharmacology , Patch-Clamp Techniques , Propofol , Random Allocation , Receptor, Muscarinic M3/drug effects , Receptor, Muscarinic M3/physiology , Receptors, Adrenergic, alpha-1/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Reflex, Righting/drug effects , Single-Blind Method , Specific Pathogen-Free Organisms , Synaptic Potentials/drug effects , Synaptic Potentials/physiology
17.
Front Mol Neurosci ; 11: 420, 2018.
Article in English | MEDLINE | ID: mdl-30564094

ABSTRACT

General anesthesia has been used clinically for more than 170 years, yet its underlying mechanisms are still not fully understood. The parabrachial nucleus (PBN) in the brainstem has been known to be crucial for regulating wakefulness and signs of arousal on the cortical electroencephalogram (EEG). Lesions of the parabrachial complex lead to unresponsiveness and a monotonous high-voltage, and a slow-wave EEG, which are the two main features of general anesthesia. However, it is unclear whether and how the PBN functions in the process of general anesthesia. By recording the levels of calcium in vivo in real-time, we found that the neural activity in PBN is suppressed during anesthesia, while it is robustly activated during recovery from propofol and isoflurane anesthesia. The activation of PBN neurons by "designer receptors exclusively activated by designer drugs" (DREADDs) shortened the recovery time but did not change the induction time. Cortical EEG recordings revealed that the neural activation of PBN specifically affected the recovery period, with a decrease of δ-band power or an increase in ß-band power; no EEG changes were seen in the anesthesia period. Furthermore, the activation of PBN elicited neural activation in the prefrontal cortex, basal forebrain, lateral hypothalamus, thalamus, and supramammillary nucleus. Thus, PBN is critical for behavioral and electroencephalographic arousal without affecting the induction of general anesthesia.

18.
Mol Pain ; 14: 1744806918803039, 2018.
Article in English | MEDLINE | ID: mdl-30198382

ABSTRACT

Background Cancer pain is a well-known serious complication in metastatic or terminal cancer patients. Current pain management remains unsatisfactory. The activation of spinal and supraspinal P2X7 receptors plays a crucial role in the induction and maintenance mechanisms of various kinds of acute or chronic pain. The midbrain periaqueductal gray is a vital supraspinal site of the endogenous descending pain-modulating system. Tramadol is a synthetic, centrally acting analgesic agent that exhibits considerable efficacy in clinically relieving pain. The purpose of this study was to determine whether the activation of P2X7 receptor in the ventrolateral region of the periaqueductal gray (vlPAG) participates in the analgesic mechanisms of tramadol on bone cancer pain in rats. The bone cancer pain rat model was established by intratibial cell inoculation of SHZ-88 mammary gland carcinoma cells. The analgesic effects of different doses of tramadol (10, 20, and 40 mg/kg) were assessed by measuring the mechanical withdrawal threshold and thermal withdrawal latency values in rats by using an electronic von Frey anesthesiometer and radiant heat stimulation, respectively. Alterations in the number of P2X7 receptor-positive cells and P2X7 protein levels in vlPAG were separately detected by using immunohistochemistry and Western blot assay. The effect of intra-vlPAG injection of A-740003 (100 nmol), a selective competitive P2X7 receptor antagonist, on the analgesic effect of tramadol was also observed. Results The expression of P2X7 receptor in the vlPAG on bone cancer pain rats was mildly elevated, and the tramadol (10, 20, and 40 mg/kg) dose dependently relieved pain-related behaviors in bone cancer pain rats and further upregulated the expression of P2X7 receptor in the vlPAG. The intra-vlPAG injection of A-740003 pretreatment partly but significantly antagonized the analgesic effect of tramadol on bone cancer pain rats. Conclusions The injection of tramadol can dose dependently elicit analgesic effect on bone cancer pain rats by promoting the expression of the P2X7 receptor in vlPAG.


Subject(s)
Analgesics, Opioid/therapeutic use , Cancer Pain/drug therapy , Periaqueductal Gray/drug effects , Receptors, Purinergic P2X7/metabolism , Tramadol/therapeutic use , Acetamides/therapeutic use , Animals , Bone Neoplasms/complications , Bone Neoplasms/diagnostic imaging , Cancer Pain/diagnostic imaging , Cancer Pain/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hyperalgesia/drug therapy , Microinjections , Pain Measurement , Pain Threshold/drug effects , Quinolines/therapeutic use , Rats , Rats, Sprague-Dawley , Time Factors , Xenograft Model Antitumor Assays
19.
Biochimie ; 154: 119-126, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30144499

ABSTRACT

Gastric cancer (GC) is one of the most common malignancies as well as the third leading cause for cancer-related death. Molecular basis of GC are essential and critical for its therapeutic treatment, but still remain poorly understood. T-cell intracellular antigen-1 (TIA1) extensively involves in cancer progression, whereas its role and regulation mechanism in GC have not been revealed. In the present study, we found that TIA-1 protein level was down-regulated in GC tissues and TIA1 inhibited proliferation and promoted apoptosis of GC cells. Then, we used bioinformatics to predict miR-487a as the upstream regulator of TIA1 and we also observed an inverse correlation between miR-487a level and TIA-1 protein level in GC tissues. Next, we demonstrated that miR-487a directly targeted TIA1 via binding to its 3'-untranslated region. Furthermore, we investigated the role of miR-487a-TIA1 pathway in the growth of GC cells both in vitro and in vivo. The repression of TIA-1 by miR-487a promoted cell proliferation and suppressed cell apoptosis in vitro, and the knockdown of miR-487a had the opposite effects. Finally, we demonstrated that miR-487a promoted the development of gastric tumor growth in xenograft mice by targeting TIA-1. These effects could be partially reversed by restoring the expression of TIA-1. Overall, our results reveal that TIA1 is a tumor suppressor gene and is directly regulated by miR-487a in GC, which may offer new therapeutic targets for GC treatment.


Subject(s)
MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Neoplasm/metabolism , Stomach Neoplasms/metabolism , T-Cell Intracellular Antigen-1/metabolism , Animals , Female , Humans , Male , Mice , Mice, SCID , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , T-Cell Intracellular Antigen-1/genetics
20.
Front Pharmacol ; 9: 1516, 2018.
Article in English | MEDLINE | ID: mdl-30666206

ABSTRACT

Xiaochaihutang (XCHT) is one of classic prescriptions in Treatise on Febrile Diseases in China which was reported to have the effect of anti-hepatic fibrosis in vivo. Activation of hepatic stellate cells (HSCs) is now well established as a central driver of fibrosis in liver injury. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important element for anti-oxidative damage which is one of the key factors responsible for occurrence. This study was to investigate the effect of XCHT compound serum on HSCs activation and focus on the Nrf2 pathway. Rats in treatment groups were given the appropriate doses of XCHT granules (5 g/kg) and Silybin (50 mg/kg) for 6 days, and the serum were obtained. The compound serum was used to intervene HSCs. The results found that XCHT compound serum significantly inhibited the proliferation of HSCT6 cells. The number of α-SMA positive stained cells in HSCT6 cells and the content of Collagen type I (collagen-I) in supernatant were significantly decreased indicating suppression of activated HSCs. Compared with the control group, the nuclear transcription of Nrf2 and the expressions of Nqo1, GCLC, and GCLM were significantly increased in XCHT group. However, the effects of XCHT were inhibited in Nrf2-siRNA transfected HSCT6 cells. These studies demonstrated that XCHT could inhibit HSCT6 cell proliferation and activation. The mechanism might be related to up-regulation of the Nrf2 pathway against oxidative stress.

SELECTION OF CITATIONS
SEARCH DETAIL
...