Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
BMC Anesthesiol ; 24(1): 37, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38263038

ABSTRACT

OBJECTIVE: This study was aimed to explore the protective effect of electroacupuncture (EA) pretreatment at Zusanli point (ST36) on ventilation-induced lung injury (VILI) and its potential anti-inflammatory mechanism. METHODS: High tidal volume ventilation was used to induce the VILI in mice, and EA pretreatment at ST36 was given for 7 consecutive days. The wet/dry ratio and pathological injury score of lung tissue, and total protein content of pulmonary alveolar lavage fluid (BALF) were detected after 4 h of mechanical ventilation (MV). Meanwhile, the expressions of TLR4 and NF- κB in lung tissue were evaluated by Western Blot, and the inflammatory factors in lung tissue were detected by ELISA. RESULTS: After four hours of mechanical ventilation, mice with ventilator-induced lung injury showed significant increases in lung wet/dry ratio, tissue damage scores, and protein content in bronchoalveolar lavage fluid. Pro-inflammatory cytokines (IL-6, IL-1ß, TNF-α) and TLR4/NF-κB expression levels in the lung were also markedly elevated (P < 0.05). Conversely, ST36 acupuncture point pre-treatment significantly reduced these parameters (P < 0.05). CONCLUSION: EA pretreatment at ST36 could alleviate the inflammatory response for VILI via inhibiting TLR4/NF- κB pathway.


Subject(s)
Electroacupuncture , Ventilator-Induced Lung Injury , Animals , Mice , NF-kappa B , Toll-Like Receptor 4 , Signal Transduction
2.
Carcinogenesis ; 44(8-9): 682-694, 2023 12 02.
Article in English | MEDLINE | ID: mdl-37294054

ABSTRACT

EphB6 belongs to the receptor tyrosine kinase, whose low expression is associated with shorter survival of colorectal cancer (CRC) patients. But the role and mechanism of EphB6 in the progression of CRC need further study. In addition, EphB6 was mainly expressed in intestinal neurons. But how EphB6 is involved in functions of intestinal neurons has not been known. In our study, we constructed a mouse xenograft model of CRC by injecting CMT93 cells into the rectum of EphB6-deficient mice. We found that the deletion of EphB6 in mice promoted tumor growth of CMT93 cells in a xenograft model of CRC, which was independent of changes in the gut microbiota. Interestingly, inhibition of intestinal neurons by injecting botulinum toxin A into rectum of EphB6-deficient mice could eliminate the promotive effect of EphB6 deficiency on tumor growth in the xenograft model of CRC. Mechanically, the deletion of EphB6 in mice promoted the tumor growth in CRC by increasing GABA in the tumor microenvironment. Furthermore, EphB6 deficiency in mice increased the expression of synaptosomal-associated protein 25 in the intestinal myenteric plexus, which mediated the release of GABA. Our study concluded that EphB6 knockout in mice promotes tumor growth of CMT93 cells in a xenograft model of CRC by modulating GABA release. Our study found a new regulating mechanism of EphB6 on the tumor progression in CRC that is dependent on intestinal neurons.


Subject(s)
Cell Communication , Colorectal Neoplasms , Humans , Animals , Mice , Colorectal Neoplasms/metabolism , Intestines/pathology , Neurons/metabolism , Neurons/pathology , gamma-Aminobutyric Acid , Tumor Microenvironment
3.
Mol Psychiatry ; 27(2): 873-885, 2022 02.
Article in English | MEDLINE | ID: mdl-34642458

ABSTRACT

Long-term potentiation (LTP) in the hippocampus is the most studied form of synaptic plasticity. Temporal integration of synaptic inputs is essential in synaptic plasticity and is assumed to be achieved through Ca2+ signaling in neurons and astroglia. However, whether these two cell types play different roles in LTP remain unknown. Here, we found that through the integration of synaptic inputs, astrocyte inositol triphosphate (IP3) receptor type 2 (IP3R2)-dependent Ca2+ signaling was critical for late-phase LTP (L-LTP) but not early-phase LTP (E-LTP). Moreover, this process was mediated by astrocyte-derived brain-derived neurotrophic factor (BDNF). In contrast, neuron-derived BDNF was critical for both E-LTP and L-LTP. Importantly, the dynamic differences in BDNF secretion play a role in modulating distinct forms of LTP. Moreover, astrocyte- and neuron-derived BDNF exhibited different roles in memory. These observations enriched our knowledge of LTP and memory at the cellular level and implied distinct roles of astrocytes and neurons in information integration.


Subject(s)
Astrocytes , Brain-Derived Neurotrophic Factor , Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Neurons/metabolism
4.
J Transl Med ; 20(1): 218, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35562743

ABSTRACT

BACKGROUND: Early diagnosis and treatment of chronic pancreatitis (CP) are limited. In this study, St13, a co-chaperone protein, was investigated whether it constituted a novel regulatory target in CP. Meanwhile, we evaluated the value of micro-PET/CT in the early diagnosis of CP. METHODS: Data from healthy control individuals and patients with alcoholic CP (ACP) or non-ACP (nACP) were analysed. PRSS1 transgenic mice (PRSS1Tg) were treated with ethanol or caerulein to mimic the development of ACP or nACP, respectively. Pancreatic lipid metabolite profiling was performed in human and PRSS1Tg model mice. The potential functions of St13 were investigated by crossing PRSS1Tg mice with St13-/- mice via immunoprecipitation and lipid metabolomics. Micro-PET/CT was performed to evaluate pancreatic morphology and fibrosis in CP model. RESULTS: The arachidonic acid (AA) pathway ranked the most commonly dysregulated lipid pathway in ACP and nACP in human and mice. Knockout of St13 exacerbated fatty replacement and fibrosis in CP model. Sdf2l1 was identified as a binding partner of St13 as it stabilizes the IRE1α-XBP1s signalling pathway, which regulates COX-2, an important component in AA metabolism. Micro-PET/CT with 68Ga-FAPI-04 was useful for evaluating pancreatic morphology and fibrosis in CP model mice 2 weeks after modelling. CONCLUSION: St13 is functionally activated in acinar cells and protects against the cellular characteristics of CP by binding Sdf2l1, regulating AA pathway. 68Ga-FAPI-04 PET/CT may be a very valuable approach for the early diagnosis of CP. These findings thus provide novel insights into both diagnosis and treatment of CP.


Subject(s)
Acinar Cells , Endoribonucleases , Animals , Humans , Mice , Acinar Cells/metabolism , Arachidonic Acid/metabolism , Carrier Proteins/metabolism , Endoribonucleases/metabolism , Fibrosis , Gallium Radioisotopes , Mice, Knockout , Positron Emission Tomography Computed Tomography , Protein Serine-Threonine Kinases , Trypsin/metabolism , Tumor Suppressor Proteins/metabolism
5.
Toxicol Appl Pharmacol ; 360: 109-119, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30282042

ABSTRACT

Nux vomica has been effectively used in Traditional Chinese Medicine. The processing of Nux vomica is necessary to reduce toxicity before it can be used in clinical practice. However, the mechanism for processing detoxification is unclear. hERG channels have been subjected to a routine test for compound cardiac toxicity in the drug development process. Therefore, we examined the effects and mechanisms of strychnine and brucine, two main ingredients of Nux vomica, and their N-oxides on hERG channels. Strychnine and brucine exhibited concentration-dependent inhibition of hERG channels with IC50 values of 25.9 µM and 44.18 µM, respectively. However, their nitrogen oxidative derivatives produced by processing of Nux vomica, strychnine N-oxide and brucine N-oxide, lost their activity on hERG channels. Compared to their parent compounds, only an oxygen atom was introduced in the nitrogen oxidative isoforms to compensate for the N+ - charge, suggesting that the protonated nitrogen is the key group for strychnine and brucine binding to hERG channel. Alanine-mutagenesis identified Y652 is the most important residue for strychnine and brucine binding to hERG channel. Y652A mutation increased the IC50 for strychnine and brucine by 21.64-fold and 29.78-fold that of WT IhERG, respectively. Docking simulations suggested that the protonated nitrogen of strychnine and brucine formed a cation-π interaction with the aromatic ring of Y652. This study suggests that introduction of an oxygen to compensate for the N+ - charge could be a useful strategy for reducing hERG potency and increasing the safety margin of alkaloid-type compounds in drug development.


Subject(s)
Oxygen/metabolism , Potassium Channels/metabolism , Strychnine/analogs & derivatives , Strychnine/metabolism , Alkaloids/metabolism , Cell Line , HEK293 Cells , Humans , Medicine, Chinese Traditional/methods , Nitrogen/metabolism , Sodium/metabolism , Structure-Activity Relationship , Strychnos nux-vomica/chemistry , Transcriptional Regulator ERG/metabolism
6.
Med Sci Monit ; 23: 4334-4342, 2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28885995

ABSTRACT

BACKGROUND Glucagon-like peptide-1 (GLP-1) has been reported to exert some beneficial effects on the central nervous system (CNS). However, the effect of GLP-1 on cognitive impairment associated with type 2 diabetes is not well known. This study investigated the effect of GLP-1 on ameliorating memory deficits in type 2 diabetic rats. MATERIAL AND METHODS Type 2 diabetic rats were induced by a high-sugar, high-fat diet, followed by streptozotocin (STZ) injection and then tested in the Morris Water Maze (MWM) 1 week after the induction of diabetes. The mRNA expression of Arc, APP, BACE1, and PS1 were determined by real-time quantitative PCR, and the Arc protein was analyzed by immunoblotting and immunohistochemistry. RESULTS Type 2 diabetic rats exhibited a significant decline in learning and memory in the MWM tests, but GLP-1 treatment was able to protect this decline and significantly improved learning ability and memory. The mRNA expression assays showed that GLP-1 treatment markedly reduced Arc, APP, BACE1, and PS1 expressions, which were elevated in the diabetic rats. Immunoblotting and immunohistochemistry results also confirmed that Arc protein increased in the hippocampus of diabetic rats, but was reduced after GLP-1 treatment. CONCLUSIONS Our findings suggest that GLP-1 treatment improves learning and memory deficits in type 2 diabetic rats, and this effect is likely through the reduction of Arc expression in the hippocampus.


Subject(s)
Cognitive Dysfunction/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/pharmacology , Animals , Blood Glucose/metabolism , Cognitive Dysfunction/blood , Cognitive Dysfunction/metabolism , Cytoskeletal Proteins/metabolism , Diabetes Mellitus, Type 2/blood , Diet, High-Fat/adverse effects , Disease Models, Animal , Hippocampus/drug effects , Male , Maze Learning/drug effects , Memory Disorders/complications , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley
7.
Stem Cells ; 31(8): 1633-43, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23630193

ABSTRACT

Astrocytes are key components of the niche for neural stem cells (NSCs) in the adult hippocampus and play a vital role in regulating NSC proliferation and differentiation. However, the exact molecular mechanisms by which astrocytes modulate NSC proliferation have not been identified. Here, we identified adenosine 5'-triphosphate (ATP) as a proliferative factor required for astrocyte-mediated proliferation of NSCs in the adult hippocampus. Our results indicate that ATP is necessary and sufficient for astrocytes to promote NSC proliferation in vitro. The lack of inositol 1,4,5-trisphosphate receptor type 2 and transgenic blockage of vesicular gliotransmission induced deficient ATP release from astrocytes. This deficiency led to a dysfunction in NSC proliferation that could be rescued via the administration of exogenous ATP. Moreover, P2Y1-mediated purinergic signaling is involved in the astrocyte promotion of NSC proliferation. As adult hippocampal neurogenesis is potentially involved in major mood disorder, our results might offer mechanistic insights into this disease.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Neural Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Inositol 1,4,5-Trisphosphate Receptors/deficiency , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis , Signal Transduction
8.
Nat Commun ; 15(1): 4347, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773146

ABSTRACT

Epigenetic mechanisms bridge genetic and environmental factors that contribute to the pathogenesis of major depression disorder (MDD). However, the cellular specificity and sensitivity of environmental stress on brain epitranscriptomics and its impact on depression remain unclear. Here, we found that ALKBH5, an RNA demethylase of N6-methyladenosine (m6A), was increased in MDD patients' blood and depression models. ALKBH5 in astrocytes was more sensitive to stress than that in neurons and endothelial cells. Selective deletion of ALKBH5 in astrocytes, but not in neurons and endothelial cells, produced antidepressant-like behaviors. Astrocytic ALKBH5 in the mPFC regulated depression-related behaviors bidirectionally. Meanwhile, ALKBH5 modulated glutamate transporter-1 (GLT-1) m6A modification and increased the expression of GLT-1 in astrocytes. ALKBH5 astrocyte-specific knockout preserved stress-induced disruption of glutamatergic synaptic transmission, neuronal atrophy and defective Ca2+ activity. Moreover, enhanced m6A modification with S-adenosylmethionine (SAMe) produced antidepressant-like effects. Our findings indicate that astrocytic epitranscriptomics contribute to depressive-like behaviors and that astrocytic ALKBH5 may be a therapeutic target for depression.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Astrocytes , Depressive Disorder, Major , Mice, Knockout , Animals , Astrocytes/metabolism , AlkB Homolog 5, RNA Demethylase/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , Mice , Humans , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/genetics , Depressive Disorder, Major/pathology , Male , Female , Disease Models, Animal , Mice, Inbred C57BL , Neurons/metabolism , Stress, Psychological/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 2/genetics , Behavior, Animal , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Depression/metabolism , Depression/genetics , Adult , Synaptic Transmission , Middle Aged
9.
Neurochem Res ; 38(10): 2216-26, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23974726

ABSTRACT

The destruction of calcium homeostasis is an important factor leading to neurological diseases. Store-operated Ca(2+) (SOC) channels are essential for Ca(2+) homeostasis in many cell types. However, whether SOC channels are involved in astrocyte activation induced by lipopolysaccharide (LPS) still remains unknown. In this study, we used LPS as an exogenous stimulation to investigate the role of SOC channels in astrocyte activation. Using calcium imaging technology, we first found that SOC channels blockers, 1-[h-[3-(4-methoxyphenyl)propoxy]-4-methoxyphenethyl]-1H-imidazole (SKF-96365) and 2-aminoethyldiphenyl borate (2-APB), inhibited LPS induced [Ca(2+)]i increase, which prompted us to speculate that SOC channels may be involved in LPS induced astrocyte activation. Further experiments confirmed our speculation shown as SOC channels blockers inhibited LPS induced astrocyte activation characterized as cell proliferation by MTS and BrdU assay, raise in glial fibrillary acidic protein expression by immunofluorescence and Western Blot and secretion of interleukin 6 (IL-6) and interleukin 1ß (IL-1ß) by ELISA. So, our studies showed that SOC channels are involved in LPS-induced astrocyte activation.


Subject(s)
Astrocytes/physiology , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Animals , Astrocytes/drug effects , Boron Compounds/pharmacology , Calcium Channels/drug effects , Calcium Signaling/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Glial Fibrillary Acidic Protein/biosynthesis , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Rats , Rats, Wistar
10.
Acta Pharmacol Sin ; 34(12): 1491-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24304919

ABSTRACT

AIM: Aptamers are oligonucleic acid or peptide molecules that bind to a specific target molecule in cells, thus may act as effective vehicles for drug or siRNA delivery. In this study we investigated the DNA aptamers that target human glioblastoma multiforme (GBM) cells overexpressing epidermal growth factor receptor variant III (EGFRvIII), which was linked to radiation and chemotherapeutic resistance of this most aggressive brain tumor. METHODS: A 73-mer ssDNA library containing molecules with 30 nt of random sequence flanked by two primer hybridization sites was chosen as the initial library. Cell systematic evolution of ligands by exponential enrichment (Cell-SELEX) method was used to select the DNA aptamers that target EGFRvIII. The binding affinity of the aptamers was measured using a cell-based biotin-avidin ELISA. RESULTS: After 14 rounds of selection, four DNA aptamers (32, 41, 43, and 47) that specifically bound to the EGFRvIII-overexpressing human glioma U87Δ cells with Kd values of less than 100 nmol/L were discovered. These aptamers were able to distinguish the U87Δ cells from the negative control human glioma U87MG cells and HEK293 cells. Aptamer 32 specifically bound to the EGFRvIII protein with an affinity similar to the EGFR antibody (Kd values of aptamer 32 and the EGFR antibody were 0.62±0.04 and 0.32±0.01 nmol/L, respectively), and this aptamer was localized in the cell nucleus. CONCLUSION: The DNA aptamers are promising molecular probes for the diagnosis and treatment of GBM.


Subject(s)
Aptamers, Nucleotide/pharmacology , Brain Neoplasms/pathology , ErbB Receptors/metabolism , Glioblastoma/pathology , Base Sequence , Blotting, Western , Brain Neoplasms/metabolism , Cell Line, Tumor , DNA Primers , Flow Cytometry , Glioblastoma/metabolism , Humans , Molecular Probes , SELEX Aptamer Technique
11.
Proc Natl Acad Sci U S A ; 107(50): 21818-23, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21106764

ABSTRACT

Neuregulin 1 (NRG1) is a trophic factor that acts by stimulating ErbB receptor tyrosine kinases and has been implicated in neural development and synaptic plasticity. In this study, we investigated mechanisms of its suppression of long-term potentiation (LTP) in the hippocampus. We found that NRG1 did not alter glutamatergic transmission at SC-CA1 synapses but increased the GABA(A) receptor-mediated synaptic currents in CA1 pyramidal cells via a presynaptic mechanism. Inhibition of GABA(A) receptors blocked the suppressing effect of NRG1 on LTP and prevented ecto-ErbB4 from enhancing LTP, implicating a role of GABAergic transmission. To test this hypothesis further, we generated parvalbumin (PV)-Cre;ErbB4(-/-) mice in which ErbB4, an NRG1 receptor in the brain, is ablated specifically in PV-positive interneurons. NRG1 was no longer able to increase inhibitory postsynaptic currents and to suppress LTP in PV-Cre;ErbB4(-/-) hippocampus. Accordingly, contextual fear conditioning, a hippocampus-dependent test, was impaired in PV-Cre;ErbB4(-/-) mice. In contrast, ablation of ErbB4 in pyramidal neurons had no effect on NRG1 regulation of hippocampal LTP or contextual fear conditioning. These results demonstrate a critical role of ErbB4 in PV-positive interneurons but not in pyramidal neurons in synaptic plasticity and support a working model that NRG1 suppresses LTP by enhancing GABA release. Considering that NRG1 and ErbB4 are susceptibility genes of schizophrenia, these observations contribute to a better understanding of how abnormal NRG1/ErbB4 signaling may be involved in the pathogenesis of schizophrenia.


Subject(s)
ErbB Receptors/metabolism , Interneurons/metabolism , Long-Term Potentiation/physiology , Neuregulin-1/metabolism , Parvalbumins/metabolism , Animals , Conditioning, Psychological , ErbB Receptors/genetics , Fear , GABA-A Receptor Antagonists/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Interneurons/cytology , Long-Term Potentiation/drug effects , Mice , Mice, Knockout , Neuregulin-1/genetics , Receptor, ErbB-4 , Receptors, GABA-A/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism
12.
MycoKeys ; 99: 297-317, 2023.
Article in English | MEDLINE | ID: mdl-37899767

ABSTRACT

Species of Apiospora are distributed worldwide as endophytes, pathogens and saprobes. In this study, we analysed Apiospora strains isolated from diseased leaves in Yunnan Province and dead culms in Shaanxi Province, China and we identified fungal species based on multi-locus phylogeny of ITS, LSU, tef1 and tub2 genes, along with the morphological characters, host and ecological distribution. Analyses revealed three new species, namely A.corylisp. nov., A.lophatherisp. nov. and A.oenotheraesp. nov. and one known species A.arundinis. Illustrations and descriptions of the four taxa are provided, along with comparisons with closely-related taxa in the genus.

13.
Neuropsychopharmacology ; 48(8): 1164-1174, 2023 07.
Article in English | MEDLINE | ID: mdl-36797374

ABSTRACT

Pharmacological manipulation of mGluR5 has showed that mGluR5 is implicated in the pathophysiology of anxiety and mGluR5 has been proposed as a potential drug target for anxiety disorders. Nevertheless, the mechanism underlying the mGluR5 involvement in stress-induced anxiety-like behavior remains largely unknown. Here, we found that chronic restraint stress induced anxiety-like behavior and decreased the expression of mGluR5 in hippocampal CA1. Specific knockdown of mGluR5 in hippocampal CA1 pyramidal neurons produced anxiety-like behavior. Furthermore, both chronic restraint stress and mGluR5 knockdown impaired inhibitory synaptic inputs in hippocampal CA1 pyramidal neurons. Notably, positive allosteric modulator of mGluR5 rescued stress-induced anxiety-like behavior and restored the inhibitory synaptic inputs. These findings point to an essential role for mGluR5 in hippocampal CA1 pyramidal neurons in mediating stress-induced anxiety-like behavior.


Subject(s)
Hippocampus , Pyramidal Cells , Hippocampus/metabolism , Pyramidal Cells/physiology , Anxiety/drug therapy , CA1 Region, Hippocampal
14.
J Clin Invest ; 133(7)2023 04 03.
Article in English | MEDLINE | ID: mdl-36757814

ABSTRACT

Major depressive disorder is a common and devastating psychiatric disease, and the prevalence and burden are substantially increasing worldwide. Multiple studies of depression patients have implicated glucose metabolic dysfunction in the pathophysiology of depression. However, the molecular mechanisms by which glucose and related metabolic pathways modulate depressive-like behaviors are largely uncharacterized. Uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) is a glucose metabolite with pivotal functions as a donor molecule for O-GlcNAcylation. O-GlcNAc transferase (OGT), a key enzyme in protein O-GlcNAcylation, catalyzes protein posttranslational modification by O-GlcNAc and acts as a stress sensor. Here, we show that Ogt mRNA was increased in depression patients and that astroglial OGT expression was specifically upregulated in the medial prefrontal cortex (mPFC) of susceptible mice after chronic social-defeat stress. The selective deletion of astrocytic OGT resulted in antidepressant-like effects, and moreover, astrocytic OGT in the mPFC bidirectionally regulated vulnerability to social stress. Furthermore, OGT modulated glutamatergic synaptic transmission through O-GlcNAcylation of glutamate transporter-1 (GLT-1) in astrocytes. OGT astrocyte-specific knockout preserved the neuronal morphology atrophy and Ca2+ activity deficits caused by chronic stress and resulted in antidepressant effects. Our study reveals that astrocytic OGT in the mPFC regulates depressive-like behaviors through the O-GlcNAcylation of GLT-1 and could be a potential target for antidepressants.


Subject(s)
Astrocytes , Depressive Disorder, Major , Mice , Animals , Astrocytes/metabolism , Depression/genetics , Synaptic Transmission , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Antidepressive Agents , Glucose , Acetylglucosamine/metabolism
15.
Anal Chim Acta ; 1211: 339907, 2022 Jun 08.
Article in English | MEDLINE | ID: mdl-35589231

ABSTRACT

A three-dimensional g-C3N4/MWNTs/GO hybrid modified electrode was constructed as an electrochemical sensor for the simultaneous determination of ascorbic acid (AA), dopamine (DA) and uric acid (UA). Due to the high conductivity of MWCNTs and the strong synergy between g-C3N4 and GO, the combination of the three effectively improved the electrocatalytic activity of the modified electrode for the oxidation of AA, DA, and UA, and solved the problems such as overlapping anodic peaks. The electrochemical performance of the as-constructed sensor was investigated and optimized by cyclic voltammetry (CV) and differential pulse voltammetry (DPV). The linear response range of AA, DA, and UA in the optimal condition was 0.2-7.5 mM, 2-100 µM, and 4-200 µM, respectively. The detection limits (S/N = 3) of AA, DA, and UA were 96, 0.22, and 1.36 µM, respectively. The recoveries of AA, DA and UA in serum samples from three groups were 92.82-106.50%, and the relative standard deviations were less than 2%. The results show that the as-constructed g-C3N4/MWNTs/GO modified electrode has the advantages of simplicity, high sensitivity and good selectivity, and can simultaneously determine AA, DA, and UA.


Subject(s)
Graphite , Uric Acid , Ascorbic Acid , Dopamine , Electrochemical Techniques/methods , Electrodes
16.
Front Neurol ; 13: 821917, 2022.
Article in English | MEDLINE | ID: mdl-35669869

ABSTRACT

Status epilepticus (SE) is a medical emergency associated with acute severe systemic damage and high mortality. Moreover, symptomatic SE is one of the highest risk factors for epileptogenesis. While the antiepileptic drugs (AEDs) are chosen in favor of acute control of SE, the potential short-term and long-term effects of such AEDs have been ignored in clinics. In this study, we hypothesized that AEDs that are used to control acute SE might affect the feasibility for the chronic development of epileptogenesis after SE. Therefore, we sought to compare the epileptogenic effects of SE that are terminated by three AEDs, i.e., diazepam, midazolam, and pentobarbital, which are widely used as first-line anti-SE AEDs. For this purpose, we used a mouse model of SE induced by intraperitoneal (i.p.) injection of lithium chloride (LiCl)-pilocarpine. The pilocarpine-induced SE was terminated with diazepam, midazolam, or pentobarbital. Then we compared short-term and long-term effects of SE with different AED treatments by examining SE-associated mortality and behavioral spontaneous recurrent seizures (SRSs) and by using magnetic resonance imaging (MRI) and immunohistochemistry to evaluate pathological and cellular alterations of mice in the different treatment groups. We found that i.p. injections of diazepam (5 mg/kg), midazolam (10 mg/kg), and pentobarbital (37.5 mg/kg) were able to terminate acute pilocarpine-SE effectively, while pentobarbital treatment showed less neuroprotective action against lethality in the short phase following SE. Long-term evaluation following SE revealed that SE treated with midazolam had resulted in relatively less behavioral SRS, less hippocampal atrophy, and milder neuronal loss and gliosis. Our data revealed an obvious advantage of midazolam vs. diazepam or pentobarbital in protecting the brain from epileptogenesis. Therefore, if midazolam provides as strong action to quench SE as other AEDs in clinics, midazolam should be the first choice of anti-SE AEDs as it provides additional benefits against epileptogenesis.

17.
Theranostics ; 12(8): 3703-3718, 2022.
Article in English | MEDLINE | ID: mdl-35664080

ABSTRACT

Rationale: Stress is a major risk factor for the development of depression. However, the underlying molecular mechanisms of stress vulnerability in depression are largely uncharacterized. Methods: P2X2 receptors (a major receptor for gliotransmitter-ATP) in the medial prefrontal cortex (mPFC) were identified by real-time qPCR, western blots and RNAscope in situ hybridization in chronic social defeat stress model (CSDS). We generated P2X2 conditional knockout mice and overexpressed AAV-P2X2 in CamkIIα-Cre mice. The depression-like behaviors were assessed via CSDS, subthreshold social defeat stress (SSDS), social interaction test (SI), forced interaction test (FIT), forced swimming test (FST), sucrose preference test (SPT), novel stressed feeding (NSF) and open field test (OFT). The neuronal activity and synapse function of P2X2 receptors in the mPFC were detected by in vivo fiber-photometry, patch-clamp techniques and neuronal morphometric analysis. Results: We identified that P2X2 receptors were increased in the mPFC of susceptible mice in CSDS. Conditional knockout of P2X2 receptors in pyramidal neurons promoted resilience of chronic stress-induced depressive-like behaviors, whereas pyramidal neurons - specific gain of P2X2 in the mPFC increased vulnerability to depressive-like behaviors. In vivo fiber-photometry, electrophysiology and neuronal morphometric analysis showed P2X2 receptors regulated neuronal activity and synapse function in the mPFC. Conclusions: Overall, our studies reveal a critical role of P2X2 in mediating vulnerability to chronic stress and identify P2X2 as a potential therapeutic target for treatment of stress-related mood disorders.


Subject(s)
Pyramidal Cells , Stress, Psychological , Animals , Mice , Mice, Inbred C57BL , Neurons , Receptors, Purinergic P2X2
18.
Biol Psychiatry ; 92(3): 204-215, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35151464

ABSTRACT

BACKGROUND: Major depressive disorder is a devastating psychiatric illness that affects approximately 17% of the population worldwide. Astrocyte dysfunction has been implicated in its pathophysiology. Traumatic experiences and stress contribute to the onset of major depressive disorder, but how astrocytes respond to stress is poorly understood. METHODS: Using Western blotting analysis, we identified that stress vulnerability was associated with reduced astrocytic glucocorticoid receptor (GR) expression in mouse models of depression. We further investigated the functions of astrocytic GRs in regulating depression and the underlying mechanisms by using a combination of behavioral studies, fiber photometry, biochemical experiments, and RNA sequencing methods. RESULTS: GRs in astrocytes were more sensitive to stress than those in neurons. GR absence in astrocytes induced depressive-like behaviors, whereas restoring astrocytic GR expression in the medial prefrontal cortex prevented the depressive-like phenotype. Furthermore, we found that GRs in the medial prefrontal cortex affected astrocytic Ca2+ activity and dynamic ATP (adenosine 5'-triphosphate) release in response to stress. RNA sequencing of astrocytes isolated from GR deletion mice identified the PI3K-Akt (phosphoinositide 3-kinase-Akt) signaling pathway, which was required for astrocytic GR-mediated ATP release. CONCLUSIONS: These findings reveal that astrocytic GRs play an important role in stress response and that reduced astrocytic GR expression in the stressed subject decreases ATP release to mediate stress vulnerability.


Subject(s)
Astrocytes , Depressive Disorder, Major , Adenosine Triphosphate/metabolism , Animals , Astrocytes/metabolism , Depressive Disorder, Major/metabolism , Glucocorticoids/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Glucocorticoid/metabolism
19.
Cell Death Dis ; 13(10): 893, 2022 10 22.
Article in English | MEDLINE | ID: mdl-36273194

ABSTRACT

Noninflammatory clearance of dying cells by professional phagocytes, termed efferocytosis, is fundamental in both homeostasis and inflammatory fibrosis disease but has not been confirmed to occur in chronic pancreatitis (CP). Here, we investigated whether efferocytosis constitutes a novel regulatory target in CP and its mechanisms. PRSS1 transgenic (PRSS1Tg) mice were treated with caerulein to mimic CP development. Phospholipid metabolite profiling and epigenetic assays were performed with PRSS1Tg CP models. The potential functions of Atp8b1 in CP model were clarified using Atp8b1-overexpressing adeno-associated virus, immunofluorescence, enzyme-linked immunosorbent assay(ELISA), and lipid metabolomic approaches. ATAC-seq combined with RNA-seq was then used to identify transcription factors binding to the Atp8b1 promoter, and ChIP-qPCR and luciferase assays were used to confirm that the identified transcription factor bound to the Atp8b1 promoter, and to identify the specific binding site. Flow cytometry was performed to analyze the proportion of pancreatic macrophages. Decreased efferocytosis with aggravated inflammation was identified in CP. The lysophosphatidylcholine (LPC) pathway was the most obviously dysregulated phospholipid pathway, and LPC and Atp8b1 expression gradually decreased during CP development. H3K27me3 ChIP-seq showed that increased Atp8b1 promoter methylation led to transcriptional inhibition. Atp8b1 complementation substantially increased the LPC concentration and improved CP outcomes. Bhlha15 was identified as a transcription factor that binds to the Atp8b1 promoter and regulates phospholipid metabolism. Our study indicates that the acinar Atp8b1/LPC pathway acts as an important "find-me" signal for macrophages and plays a protective role in CP, with Atp8b1 transcription promoted by the acinar cell-specific transcription factor Bhlha15. Bhlha15, Atp8b1, and LPC could be clinically translated into valuable therapeutic targets to overcome the limitations of current CP therapies.


Subject(s)
Adenosine Triphosphatases , Lysophosphatidylcholines , Macrophages , Pancreatitis, Chronic , Animals , Mice , Acinar Cells/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Ceruletide/toxicity , Histones/metabolism , Inflammation/metabolism , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Macrophages/metabolism , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Transcription Factors/metabolism
20.
J Neurosci ; 30(38): 12653-63, 2010 Sep 22.
Article in English | MEDLINE | ID: mdl-20861371

ABSTRACT

Increasing evidence indicates that stimulating hippocampal neurogenesis could provide novel avenues for the treatment of depression, and recent studies have shown that in vitro neurogenesis is enhanced by hypoxia. The aim of this study was to investigate the potential regulatory capacity of an intermittent hypobaric hypoxia (IH) regimen on hippocampal neurogenesis and its possible antidepressant-like effect. Here, we show that IH promotes the proliferation of endogenous neuroprogenitors leading to more newborn neurons in hippocampus in adult rats. Importantly, IH produces antidepressant-like effects in multiple animal models screening for antidepressant activity, including the forced swimming test, chronic mild stress paradigm, and novelty-suppressed feeding test. Hippocampal x-ray irradiation blocked both the neurogenic and behavioral effects of IH, indicating that IH likely produces antidepressant-like effects via promoting neurogenesis in adult hippocampus. Furthermore, IH stably enhanced the expression of BDNF in hippocampus; both the antidepressant-like effect and the enhancement of cell proliferation induced by IH were totally blocked by pharmacological and biological inhibition of BDNF-TrkB (tyrosine receptor kinase B) signaling, suggesting that the neurogenic and antidepressant-like effects of IH may involve BDNF signaling. These observations might contribute to both a better understanding of physiological responses to IH and to developing IH as a novel therapeutic approach for depression.


Subject(s)
Hippocampus/physiology , Hypoxia/metabolism , Motor Activity/physiology , Neurogenesis/physiology , Analysis of Variance , Animals , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/metabolism , Fluoxetine/pharmacology , Hippocampus/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Male , Motor Activity/drug effects , Neurogenesis/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Stress, Physiological/drug effects , Stress, Physiological/physiology , Stress, Psychological/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL