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










Publication year range
1.
Int Immunopharmacol ; 133: 112157, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38678671

ABSTRACT

In non-small cell lung cancer (NSCLC), identifying a component with certain molecular targets can aid research on cancer treatment. Dihydroartemisinin (DHA) is a semisynthetic derivative of artemisinin which induced the anti-cancer effects via the STAT3 signaling pathway, but the underlying molecular mechanism is still elusive. In this study, we first proved that DHA prohibits the growth of tumors both in vitro and in vivo. Data from transcriptomics showed that DHA reduced the expression level of the genes involved in cell cycle-promoting and anti-apoptosis, and most importantly, DHA restricted the expression level of receptor tyrosine kinase-like orphan receptor 1 (ROR1) which has been reported to have abnormal expression on tumor cells and had close interaction with STAT3 signaling. Then, we performed comprehensive experiments and found that DHA remarkably decreased the expression of ROR1 at both mRNA and protein levels and it also diminished the phosphorylation level of STAT3 in NSCLC cell lines. In addition, our data showed that exogenously introduced ROR1 could significantly enhance the phosphorylation of STAT3 while blocking ROR1 had the opposite effects indicating that ROR1 plays a critical role in promoting the activity of STAT3 signaling. Finally, we found that ROR1 overexpression could partially reverse the decreased activity of STAT3 induced by DHA which indicates that DHA-induced anti-growth signaling is conferred, at least in part, through blocking ROR1-mediated STAT3 activation. In summary, our study indicates that in NSCLC, ROR1 could be one of the critical molecular targets mediating DHA-induced STAT3 retardation.


Subject(s)
Artemisinins , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Receptor Tyrosine Kinase-like Orphan Receptors , STAT3 Transcription Factor , Artemisinins/pharmacology , Artemisinins/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Animals , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Mice, Nude , Apoptosis/drug effects , Mice , Gene Expression Regulation, Neoplastic/drug effects , A549 Cells , Mice, Inbred BALB C
2.
Int Immunopharmacol ; 123: 110686, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37499397

ABSTRACT

Over the last decade, immuno-oncologic drugs especially CD3-engaging bispecific antibodies (biAbs) are experiencing fast-paced evolution, but big challenges still exist in the clinical development of biAbs in solid tumors, especially non-small cell lung cancer (NSCLC). In this study, we choose a ROR1 × CD3 biAb in scFv-Fc format, named R11 × v9 biAb, to investigate its tumor-inhibiting role in NSCLC. Notably, the ROR1-engaging arm binds both human and mouse ROR1. We found that R11 × v9 biAb specifically binds T cells and tumor cells simultaneously, and dose-dependent cytotoxicity was detected for various ROR1+ NSCLC cell lines. Further, R11 × v9 biAb mediated T-cell derived proinflammatory cytokine secretion, boosted granzyme B and perforin production from CD8+ T cells, and recruited more CD4+ T cells and CD8+ T cells into the tumor tissues. The antitumor activity of R11 × v9 biAb was confirmed in two xenograft mouse models of ROR1+ NSCLC. Importantly, no harmful side effects were observed in these in vivo studies, warranting further preclinical and clinical studies of R11 × v9 biAb in NSCLC.


Subject(s)
Antibodies, Bispecific , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Humans , Mice , Carcinoma, Non-Small-Cell Lung/immunology , CD3 Complex , CD8-Positive T-Lymphocytes , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Receptor Tyrosine Kinase-like Orphan Receptors
3.
Am J Chin Med ; 51(5): 1127-1151, 2023.
Article in English | MEDLINE | ID: mdl-37335209

ABSTRACT

The brain metabolic changes caused by the interruption of blood supply are the initial factors of brain injury in ischemic stroke. Electroacupuncture (EA) pretreatment has been shown to protect against ischemic stroke, but whether its neuroprotective mechanism involves metabolic regulation remains unclear. Based on our finding that EA pretreatment significantly alleviated ischemic brain injury in mice by reducing neuronal injury and death, we performed a gas chromatography-time of flight mass spectrometry (GC-TOF/MS) to investigate the metabolic changes in the ischemic brain and whether EA pretreatment influenced these changes. First, we found that some glycolytic metabolites in the normal brain tissues were reduced by EA pretreatment, which may lay the foundation of neuroprotection for EA pretreatment against ischemic stroke. Then, 6[Formula: see text]h of cerebral ischemia-induced brain metabolic changes, especially the enhanced glycolysis, were partially reversed by EA pretreatment, which was manifested by the brain levels of 11 of 35 up-regulated metabolites and 18 of 27 down-regulated metabolites caused by cerebral ischemia significantly decreasing and increasing, respectively, due to EA pretreatment. A further pathway analysis showed that these 11 and 18 markedly changed metabolites were mainly involved in starch and sucrose metabolism, purine metabolism, aspartate metabolism, and the citric acid cycle. Additionally, we found that EA pretreatment raised the levels of neuroprotective metabolites in both normal and ischemic brain tissues. In conclusion, our study revealed that EA pretreatment may attenuate the ischemic brain injury by inhibiting glycolysis and increasing the levels of some neuroprotective metabolites.


Subject(s)
Brain Injuries , Brain Ischemia , Electroacupuncture , Ischemic Stroke , Reperfusion Injury , Stroke , Mice , Animals , Electroacupuncture/methods , Neuroprotection , Brain Ischemia/metabolism , Metabolomics , Reperfusion Injury/prevention & control , Stroke/etiology , Stroke/prevention & control
4.
Behav Brain Res ; 441: 114287, 2023 03 12.
Article in English | MEDLINE | ID: mdl-36627054

ABSTRACT

Lack of maternal care and attention during infancy and childhood increases the likelihood of developing a range of neuropsychiatric disorders, such as social deficits, working memory impairment, and anxiety-like behaviors, in adulthood. However, the neuroregulatory signaling through which early-life stress causes behavioral and cognitive abnormalities in the offspring is largely unexplored. Here, we show that in mice, unpredictable maternal separation (MS) during the early postnatal period impairs neuronal development in the medial prefrontal cortex (mPFC) and results in long-lasting behavioral changes. Additionally, MS disrupts excitatory neurotransmission and inhibits the neuronal activity of pyramidal neurons in the mPFC. Differentially expressed gene (DEG) analysis of RNA sequencing (RNA-seq) data of mPFC showed that dopamine D1 receptor (D1R) was significantly downregulated in MS animals. Finally, we show that pharmacological activation of D1R signaling specifically in the mPFC improves neuronal excitability and rescues behavioral and cognitive dysfunction of MS mice, whereas pharmacologically inhibiting of D1R in the mPFC mimics MS-induced behavioral abnormalities in control mice. Together, our results identify D1R signaling in the mPFC, at least in part, as a potential therapeutic target for the behavioral and cognitive abnormalities caused by deprivation of maternal care in early life.


Subject(s)
Maternal Deprivation , Prefrontal Cortex , Mice , Animals , Prefrontal Cortex/metabolism , Synaptic Transmission , Neurons/metabolism , Receptors, Dopamine D1/metabolism
5.
Phytomedicine ; 108: 154507, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36334391

ABSTRACT

BACKGROUND: Transdermal patches are an effective form of treatment for rheumatoid arthritis (RA), and they have a number of benefits, including patient compliance, accessibility, and low systemic toxicity. ShexiangZhuifeng Analgesic Plaster (SZAP), a patch made up of many traditional medicines, has been successfully utilized in numerous clinical trials to treat RA. However, information about anti-RA processes and transdermal active components is still emerging. PURPOSE: Our objectives were to identify the transdermal active components of SZAP and investigate its anti-RA mechanisms, primarily focused on joint inflammation. METHODS: The collagen-induced arthritis (CIA) rats were created first, and then the arthritis score, Paw thickness, and morphology feature of joint synovial were assessed after 7 days of therapy with SZAP. Moreover, the Franz diffusion cell and UPLC-MS technologies were combined to identify and measure the transdermal active ingredients of SZAP. Furthermore, network pharmacology was utilized to anticipate the putative the mechanism of SZAP for treating RA. Finally, the results of network pharmacology were validated using LPS-induced RAW 264.7 cells and CIA rats. RESULTS: SZAP significantly reduced paw thickness, arthritic score and pathological characteristics of joint synovitis in (CIA) rats. Additionally, 12 transdermal active components of SZAP were identified, and network pharmacology prediction results suggested that SZAP may alleviate joint synovial inflammation by blocking the Akt/mTOR/HIF-1 pathway. Our investigations' findings demonstrated that SZAP dramatically reduced the concentrations of excess cytokines (IL6, VEGF, and TNF-α), as well as the protein overexpression of the AKT/mTOR/HIF- pathway (HIF-1, p-AKT, and p-mTOR), whereas its anti-inflammation effect was reversed once AKT or mTOR was activated. CONCLUSION: By blocking the AKT/mTOR/HIF-1 pathway, SZAP can lessen the release of inflammatory mediators, which reduces joint synovial inflammation associated with RA. The pharmacological evaluation of TCM transdermal drug delivery formulations like SZAP may be amenable to the integration of transdermal chemistry and network pharmacology approaches.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Rats , Animals , Proto-Oncogene Proteins c-akt , Chromatography, Liquid , Network Pharmacology , Tandem Mass Spectrometry , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Arthritis, Experimental/pathology , TOR Serine-Threonine Kinases , Inflammation/drug therapy , Analgesics/therapeutic use
6.
J Integr Neurosci ; 22(6): 168, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38176945

ABSTRACT

BACKGROUND: The purpose of this study was to investigate the potential involvement of pyruvate kinase M2 (PKM2), an enzyme acting as a rate-limiting enzyme in the final phase of glycolysis, in the regulation of glial activation and brain damage of intracerebral hemorrhage (ICH). METHODS: Western blotting and immunofluorescence were performed to investigate PKM2 expression, terminal deoxynucleotidyl transferase deoxyurinary triphosphate (dUTP) nick end labeling staining, hematoxylin and eosin staining, and behavioral tests were employed to evaluate the brain damage of ICH mice, and RNA-seq and bioinformatic analyses were performed to detect gene expression changes in ICH mice treated with TEPP-46. RESULTS: Increased PKM2 levels in perihematomal brain tissue were found starting from 3 days following ICH and peaked at 5 and 7 days post ICH. The increased expression of PKM2 was mainly co-localized with glial fibrillary acidic protein (GFAP)+ astrocytes and ionized calcium binding adaptor molecule-1 (IBA-1)+ microglia. Furthermore, we observed a notable increase in the nuclear translocation of PKM2 in glial cells following ICH. TEPP-46 treatment significantly reduced PKM2 nuclear translocation, and effectively attenuated glial activation and brain injury, and improved functional recovery of mice with ICH. RNA-seq data indicated that 91.1% (205/225) of differentially expressed genes (DEGs) were down-regulated in the TEPP-46 treated groups compared with the vehicle-treated groups in ICH brains. Furthermore, bioinformatic analyses revealed that these down-regulated DEGs were involved in a variety of biological processes, including autophagy and metabolic processes. In addition, the majority of these downregulated DEGs had a primary high expression in neurons, with subsequent expression seen in endothelial cells, microglia, and astrocytes. CONCLUSIONS: These results indicate that increased PKM2 nuclear translocation promotes the activation of glial cells after ICH, hence aggravating ICH-induced brain damage, and aggravates the brain injury induced by ICH. This highlights a potential therapeutic target for inhibiting glial activation to attenuate brain injury after ICH.


Subject(s)
Brain Injuries , Cerebral Hemorrhage , Neuroglia , Pyruvate Kinase , Animals , Mice , Brain Injuries/metabolism , Cerebral Hemorrhage/metabolism , Endothelial Cells/metabolism , Neuroglia/metabolism , Pyruvate Kinase/metabolism
7.
Biomed Pharmacother ; 156: 113902, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36279723

ABSTRACT

Obesity, a growing health problem in the world, is related to a series of mental disorders, including anxiety and depression. XiaoYao San (XYS), a prescription of traditional Chinese medicine (TCM), has been widely used in the clinical treatment of anxiety and depression in China. However, the efficacy of XYS on obesity-related neuropsychiatric dysfunction and the underlying neural mechanisms remain unclear. Here, using a high-fat diet (HFD)-induced obese model, we found that XYS treatment significantly improves obesity-related anxiety- and depression-like behaviors and alters the gut microbiome, particularly by increasing the relative abundance of Faecalibaculum rodentium (F. rodentium), in mice. Interestingly, selective supplementation with F. rodentium or its metabolic products, short-chain fatty acids (SCFAs), is sufficient to rescue anxiety- and depression-like behaviors in HFD-fed mice. Next, we determined that the transcriptional level of dopamine D2 receptor (DRD2), which activation usually inhibits inflammation in the central nervous system (CNS), is significantly increased in the medial prefrontal cortex (mPFC) of XYS-treated mice when compared with that of vehicle-treated controls. Moreover, enriched pathways analysis with the differential expression genes (DEGs) showed that some of these DEGs are enriched in neuroinflammatory pathways. We further noticed that treatment with XYS contributes to controlling microglial activation and proinflammatory responses in the mPFC and hippocampus of HFD-fed mice. Overall, this study reveals that XYS rescues HFD-induced anxiety and depression via modulating gut microbiota-derived metabolites and that XYS is a potential therapeutic strategy for treating obesity-associated mental disorders.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Mice , Animals , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/physiology , Depression/drug therapy , Mice, Inbred C57BL , Anxiety/drug therapy , Obesity/complications , Obesity/drug therapy , Obesity/chemically induced
8.
Colloids Surf B Biointerfaces ; 218: 112773, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36007312

ABSTRACT

Chitosan-coated poly (methacrylic acid) (PMAA) hollow spheres with 64 ± 3% drug loading capacity and low drug leakage (7 ± 2%, 54 h) were prepared through a novel one-pot two-step self-assembly process. Site-specific doxorubicin (DOX) loading and chitosan coating were achieved by electrostatic interaction to fulfill efficient drug loading and well-controlled drug release behavior. In vitro drug release profile revealed the pH and glutathione (GSH) dual responsive fast triggered drug release behavior, reaching 62 ± 3% during the first 10 h. And completely drug release could be achieved in 54 h. The high drug content and sensitive tumor microenvironment responsibility lead to similar anti-cancer efficiency with free doxorubicin in in vitro MTT assay. This self-assembly guided one-pot two-step fabrication process was proved to be an effective and convenient way to prepare the well-defined multi-layer structure and might be further employed in fabricating high-performance drug delivery systems.


Subject(s)
Chitosan , Nanoparticles , Chitosan/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Delivery Systems , Drug Liberation , Glutathione , Hydrogen-Ion Concentration , Nanoparticles/chemistry
9.
Biochem Pharmacol ; 198: 114941, 2022 04.
Article in English | MEDLINE | ID: mdl-35149053

ABSTRACT

Non-small cell lung cancer (NSCLC) cells exhibit aberrant metabolism characterized by high glycolysis even in the presence of abundant oxygen. Inhibition of aerobic glycolysis remains challenging when identifying potential cancer-specific inhibitors while maintaining or even boosting the anti-cancer immunity. Artemisinin derivatives DHA and AS have shown excellent anti-tumor and immunoenhancing roles in numerous malignancies, but the molecular mechanism of DHA and AS in regulating cancer glucose metabolism is largely unknown. In this study, we proved that DHA and AS inhibit NSCLC growth via prohibiting cancer cell aerobic glycolysis through ERK/c-Myc pathway. First, we proved that DHA and AS have comparable anti-cancer growth roles in both NSCLC cell lines and mouse Lewis Lung Cancer model. Then, our data clearly showed that DHA and AS dose- and time-dependently reduce the uptake of glucose, the production of ATP, and the secretion of lactate, the expression of glucose transporter GLUT1 and two key glycolysis-related enzymes hexokinase and lactate dehydrogenase, as well as the level of c-Myc. Finally, we generated c-Mychigh stable-expressing NSCLC cell line and treated it with DHA or AS, respectively. Our data clearly showed that c-Myc overexpression can partially reverse the glycolysis-repressing role of DHA and AS which strongly supported our proposal that AS and DHA suppress aerobic glycolysis in a c-Myc-dependent manner in NSCLC cells. This study extends our knowledge of artemisinin derivatives in regulating tumor glucose metabolism and provides potential strategy in the therapy of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Artemisinins , Artesunate , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Glucose/metabolism , Glycolysis , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
10.
Cell Mol Neurobiol ; 42(8): 2791-2804, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34460038

ABSTRACT

It is unclear how Toll-like receptor (TLR) 4 signaling affects protein succinylation in the brain after intracerebral hemorrhage (ICH). Here, we constructed a mouse ICH model to investigate the changes in ICH-associated brain protein succinylation, following a treatment with a TLR4 antagonist, TAK242, using a high-resolution mass spectrometry-based, quantitative succinyllysine proteomics approach. We characterized the prevalence of approximately 6700 succinylation events and quantified approximately 3500 sites, highlighting 139 succinyllysine site changes in 40 pathways. Further analysis showed that TAK242 treatment induced an increase of 29 succinyllysine sites on 28 succinylated proteins and a reduction of 24 succinyllysine sites on 23 succinylated proteins in the ICH brains. TAK242 treatment induced both protein hypersuccinylations and hyposuccinylations, which were mainly located in the mitochondria and cytoplasm. GO analysis showed that TAK242 treatment-induced changes in the ICH-associated succinylated proteins were mostly located in synapses, membranes and vesicles, and enriched in many cellular functions/compartments, such as metabolism, synapse, and myelin. KEGG analysis showed that TAK242-induced hyposuccinylation was mainly linked to fatty acid metabolism, including elongation and degradation. Moreover, a combined analysis of the succinylproteomic data with previously published transcriptome data revealed that most of the differentially succinylated proteins induced by TAK242 treatment were mainly distributed throughout neurons, astrocytes, and endothelial cells, and the mRNAs of seven and three succinylated proteins were highly expressed in neurons and astrocytes, respectively. In conclusion, we revealed that several TLR4 signaling pathways affect the succinylation processes and pathways in mouse ICH brains, providing new insights on the ICH pathophysiological processes. Data are available via ProteomeXchange with identifier PXD025622.


Subject(s)
Endothelial Cells , Toll-Like Receptor 4 , Animals , Brain/metabolism , Cerebral Hemorrhage/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Fatty Acids , Mice , Sulfonamides , Toll-Like Receptor 4/metabolism
11.
Eur J Pharmacol ; 908: 174338, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34270984

ABSTRACT

Neuropsychiatric dysfunction and reactive microglia are hallmarks of high-fat diet (HFD)-induced obesity, yet whether these reactive microglia contribute to HFD-induced obesity-related behavioral abnormalities and the underlying mechanisms remain unclear. Here, we show that HFD feeding causes social deficits and anxiety-like behaviors with impaired neuronal activity and alters the gut microbiota, particularly by depleting Lactobacillus reuteri (L. reuteri), in mice. The profiles of microbiome and metabolome in HFD-fed mice predict that specific microbial taxa and their metabolites regulate HFD-induced obesity-related behavioral abnormalities. Oral treatment with the L. reuteri reduces microglial activation and increases dendritic spine density, thus ameliorates social deficits and anxiety in HFD-fed mice. HFD-fed mice that are administered L. reuteri are also found to accumulate butyrate in their gut, sera and brain. Moreover, supplementation of butyrate improves behavioral abnormalities and modulates microglial homeostasis in HFD-fed mice. In addition, selectively removal of microglia through a pharmacologic approach can rescue dendritic spine loss and increase neuronal activity that profoundly alleviates social deficits and anxiety arising from HFD-induced obesity. Overall, this study reveals an unexpected pivotal role of gut commensal-derived butyrate in HFD-induced social deficits and anxiety-like behaviors through regulation of microglial homeostasis and identifies a potential probiotic treatment for HFD-induced obesity-related behavioral abnormalities.


Subject(s)
Butyrates , Microglia , Animals , Diet, High-Fat , Gastrointestinal Microbiome , Mice , Obesity
12.
Exp Ther Med ; 21(5): 426, 2021 May.
Article in English | MEDLINE | ID: mdl-33747165

ABSTRACT

Autophagy serves an important role in amyloid-ß (Aß) metabolism and τ processing and clearance in Alzheimer's disease. The progression of Aß plaque accumulation and hyperphosphorylation of τ proteins are enhanced by oxidative stress. A hydrogen peroxide (H2O2) injury cell model was established using SH-SY5Y cells. Cells were randomly divided into normal, H2O2 and chlorogenic acid (5-caffeoylquinic acid; CGA) groups. The influence of CGA on cell viability was evaluated using a Cell Counting Kit-8 assay and cell death was assessed using Hoechst 33342 nuclear staining. Autophagy induction and fusion of autophagic vacuoles assays were performed using monodansylcadaverine staining. Additionally, SH-SY5Y cells expressing Ad-mCherry-green fluorescent protein-LC3B were established to detect autophagic flow. LysoTracker Red staining was used to evaluate lysosome function and LysoSensor™ Green staining assays were used to assess lysosomal acidification. The results demonstrated that CGA decreased the apoptosis rate, increased cell viability and improved cell morphology in H2O2-treated SH-SY5Y cells. Furthermore, CGA alleviated the accumulation of autophagic vacuoles, reduced the LC3BII/I ratio and decreased P62 levels, resulting in increased autophagic flux. Additionally, CGA upregulated lysosome acidity and increased the expression levels of cathepsin D. Importantly, these effects of CGA on H2O2-treated SH-SY5Y cells were mediated via the mTOR-transcription factor EB signaling pathway. These results indicated that CGA protected cells against H2O2-induced oxidative damage via the upregulation of autophagosomes, which promoted autophagocytic degradation and increased autophagic flux.

13.
Brain Res Bull ; 166: 161-171, 2021 01.
Article in English | MEDLINE | ID: mdl-33279588

ABSTRACT

Social experiences during early life are thought to be critical for proper social and emotional development. Conversely, social insults during development causes long-lasting behavioral abnormalities later in life. However, how juvenile social deprivation influences social and emotional behaviors remains poorly understood. Here, we show that juvenile social isolation induces a shift in microbial ecology that negatively impacts social and emotional behaviors in adulthood. These behavioral changes, which occur during this critical period are transferable to antibiotic pre-treated mice by fecal microbiota transplant. In addition, juvenile social isolation decreases the expression of oxytocin receptor (OXTR) in the medial prefrontal cortex (mPFC), and increases the amounts of fecal propionic acid (PA), a short-chain fatty acid derived from gut micobiota. Accordingly, infusion with an OXTR antagonist (OXTR-A, l-368,899) specifically in the mPFC or supplementation of PA both can cause social deficits and anxiety-like behaviors in group housed mice. Collectively, our findings reveal that juvenile social experience regulates prefrontal cortical OXTR expression through gut microbiota-produced PA and that is essential for normal social and emotional behaviors, thus providing a cellular and molecular context to understand the consequences of juvenile social deprivation.


Subject(s)
Anxiety/metabolism , Gastrointestinal Microbiome/physiology , Prefrontal Cortex/metabolism , Propionates/metabolism , Receptors, Oxytocin/metabolism , Social Isolation , Animals , Behavior, Animal , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
14.
Phytother Res ; 35(3): 1572-1584, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33111362

ABSTRACT

Ligustilide is a phenolic compound isolated from Asian plants of Umbelliferae family. This study was aimed at exploring the neuroprotective effects of Ligustilide from the perspective of endoplasmic reticulum stress (ERS) and autophagy. The Alzheimer's disease (AD) cell models were constructed by SH-SY5Y cell line, which was exposed to 20 µM Aß25-35 . CCK-8 was used to evaluate the cell viability of Ligustilide on AD cell model. Hoechst staining and LysoTracker Red were used to test the cell apoptosis and Lysosome function, respectively. ERS in living cells were detected by Thioflavin T. The expression of autophagy-related proteins (LC3B-II/I, P62/SQSTM1, Beclin1, and Atg5), ERS marker proteins (PERK, GRP78, and CHOH), and apoptosis proteins (Bax, Bcl-2, and Caspase-12) were analyzed by Western blot analyses. Aß25-35 could induce ERS and autophagy in a time-dependent manner in SH-SY5Y cells. We demonstrated that Ligustilide significantly decreased the rate of apoptosis, and improved the viability of cells. Simultaneously, Ligustilide effectively modulated ERS via inhibiting the over-activation of GRP78/PERK/CHOP signaling pathway. In addition, Ligustilide alleviated the accumulation of autophagy vacuoles, reduced the ratio of LC3B-II/I and the level of P62/SQSTM1. Ligustilide significantly up-regulated lysosomal acidity and the expression of Cathepsin D (CTSD). Ligustilide could rescue lysosomal function to promote autophagy flux and inhibit the over-activation of ERS. This finding may contribute to the development of new therapeutic strategies for AD.


Subject(s)
4-Butyrolactone/analogs & derivatives , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , 4-Butyrolactone/pharmacology , 4-Butyrolactone/therapeutic use , Apoptosis , Endoplasmic Reticulum Chaperone BiP , Humans , Neuroprotective Agents/pharmacology , Signal Transduction , Transfection
15.
Behav Brain Res ; 395: 112838, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32750465

ABSTRACT

The prefrontal cortex (PFC) plays an important role in regulating anxiety-like phenotypes and social behaviors, and impairments in this brain region has been linked to social deficits in mammals. Childhood obesity is associated with an increased risk of neuropsychiatric behavioral abnormalities, including attenuated social preference and increased anxiety-like behaviors in adulthood. However, little data are available on the impact of obesity during adolescence on PFC-dependent behaviors. Herein, we use the mice pups to illuminate whether and how high-fat diet (HFD) feeding in adolescence affects medial prefrontal cortex (mPFC)-dependent behaviors, and what the underlying cellular and molecular mechanism is. We found that juvenile HFD feeding results in the accumulation of senescent astrocytes and microglia in the mPFC of mice. Furthermore, we found a causal link between the accumulation of senescent glial cells and HFD-induced neuropsychiatric behavioral abnormalities. Pharmacological clearance of senescent glial cells in HFD-fed mice enhances neuronal activity and reserves synaptic excitatory/inhibitory balance, thus preserving normal behaviors. Collectively, these results show that senescent glial cells play a significant role in the initiation and progression of juvenile obesity-mediated neuropsychiatric behavioral abnormalities, and suggest that targeting senescent glial cells may provide a therapeutic avenue for the treatment of obesity-related neuropsychiatric disorders in children.


Subject(s)
Mental Disorders/physiopathology , Neuroglia/physiology , Prefrontal Cortex/physiology , Age Factors , Aging/physiology , Animals , Anxiety/etiology , Astrocytes/physiology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/physiology , Cognition/drug effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Microglia/physiology , Neuroglia/metabolism , Neurons/physiology , Prefrontal Cortex/metabolism , Social Behavior
16.
Neuroimmunomodulation ; 27(1): 48-57, 2020.
Article in English | MEDLINE | ID: mdl-32516787

ABSTRACT

BACKGROUND AND OBJECTIVES: The enteric nervous system (ENS) dominates the onset of obesity and has been shown to regulate nutrient absorption and energy metabolism. METHODS AND STUDY DESIGN: This study was performed to investigate the role of electroacupuncture in regulating ENS function in obese mice. Obese mice were obtained by high-fat diet. 16S rRNA pyrosequencing, Western blotting, quantitative PCR, and neurotransmitter analysis were used for this purpose. RESULTS: Body weight, Lee index, serum lipid, leptin, and adiponectin levels, and other basic indices were significantly ameliorated after electroacupuncture intervention. The pathological ENS scores, serum neurotransmitter levels, and intestinal transit rate were markedly changed in obese mice. Moreover, electroacupuncture promoted the diversity of gut microbiota. No significant differences were observed 21 and 28 days after electroacupuncture. CONCLUSIONS: These results suggested ENS may be a new treatment approach to obesity.


Subject(s)
Electroacupuncture , Enteric Nervous System/physiology , Obesity/physiopathology , Animals , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/physiology , Gastrointestinal Transit/physiology , Mice , Mice, Inbred C57BL , Neurotransmitter Agents/blood
17.
Neuropsychopharmacology ; 44(12): 2054-2064, 2019 11.
Article in English | MEDLINE | ID: mdl-31207607

ABSTRACT

Obesity is one of the most serious public health challenges in the world. Obesity during early life has been associated with an increased risk of neurodevelopmental disorders, including deficits in learning and memory, yet the underlying mechanisms remain unclear. Here, we show that early life high-fat diet (HFD) feeding impairs hippocampus-dependent contextual/spatial learning and memory, and alters the gut microbiota, particularly by depleting Akkermansia muciniphila (A. muciniphila), in mice. Transplantation of the HFD microbiota confers hippocampus-dependent learning and memory deficits to mice fed a chow diet. Oral treatment of HFD-fed mice with the gut commensal A. muciniphila corrects gut permeability, reduces hippocampal microgliosis and proinflammatory cytokines (tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and IL-6) expression, and restores neuronal development and synapse plasticity, thus ameliorates defects in learning and memory. Interestingly, treatment of mice with lipopolysaccharide (LPS) mimics HFD-induced hippocampus-dependent cognitive impairment in chow-fed mice. In line with these findings, pharmacologic blockade of Toll-like receptor 4 (TLR4) signalling or antibiotics treatment both effectively prevent hippocampus-dependent learning and memory deficits in HFD-fed mice. Collectively, our findings demonstrate an unexpected pivotal role of gut microbiota in HFD-induced cognitive deficits and identify a potential probiotic therapy for obesity associated with cognitive dysfunction during early life.


Subject(s)
Cognition/physiology , Diet, High-Fat , Gastrointestinal Microbiome/physiology , Hippocampus/physiopathology , Obesity/microbiology , Obesity/physiopathology , Verrucomicrobia/physiology , Akkermansia , Animals , Conditioning, Classical/physiology , Diet, High-Fat/adverse effects , Encephalitis/microbiology , Hippocampus/growth & development , Long-Term Potentiation , Male , Mice, Inbred C57BL , Neurons/physiology , Obesity/psychology , Spatial Learning/physiology , Toll-Like Receptor 4/physiology
18.
Neural Regen Res ; 12(6): 969-976, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28761431

ABSTRACT

Synaptosomal-associated protein 25 kDa (SNAP-25) is localized on the synapse and participates in exocytosis and neurotransmitter release. Decreased expression of SNAP-25 is associated with Alzheimer's disease and attention deficit/hyperactivity disorder. However, the expression of SNAP-25 in spinal cord contusion injury is still unclear. We hypothesized that SNAP-25 is associated with sensory and locomotor functions after spinal cord injury. We established rat models of spinal cord contusion injury to detect gene changes with a gene array. A decreased level of SNAP-25 was detected by quantitative real time-polymerase chain reaction and western blot assay at 1, 3, 7, 14 and 28 days post injury. SNAP-25 was localized in the cytoplasm of neurons of the anterior and posterior horns, which are involved in locomotor and sensory functions. Our data suggest that reduced levels of SNAP-25 are associated with sensory and locomotor functions in rats with spinal cord contusion injury.

19.
Brain Res ; 1669: 106-113, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28549966

ABSTRACT

Aquaporin-4 (AQP4) is a water channel protein in spinal cord and plays a critical role in the pathophysiological process of spinal cord injury (SCI). However, little is known about the molecular mechanism of AQP4 involved in SCI. The present study was performed to investigate the possible molecules regulated by AQP4 after SCI by use of lentivirus-mediated RNA interference (RNAi). First, the motor function was evaluated by Basso, Beattie, Bresnahan (BBB) scale and the expression of AQP4 was measured by western blot, immunohistochemical staining and immunofluorescence in rats after contusion spinal cord injury (cSCI). After cSCI, the rats exhibited a gradual motor function recovery from 3dpo to 28dpo. And the expression and localization of AQP4 changed with different post-injury stages. At 3d after SCI, AQP4 located mainly in vascular endothelial cells (VECs) in the anterior horn of spinal cord, which was similar to the sham rats. At 7d and 28d after SCI, AQP4 was expressed both in VECs and the position of neuron membranes. The protein level of AQP4 increased significantly at 12h, 1d and 3d after SCI, and decreased slightly at 7d after SCI. Then lentivirus-mediated AQP4 RNA interference (AQP4-RNAi) was constructed and used to inhibit AQP4 expression in cSCI rats. The results from real-time quantitative polymerase chain reactions (RT-qPCR) and immunohistochemical staining suggested that the expression of nerve growth factor (NGF) was up-regulated by lentivirus-mediated AQP4 inhibition in cSCI rats, while the motor function recovery was accelerated. The results suggested that the acceleration of motor function recovery by AQP4 inhibition was associated with NGF up-regulation. This is the first report on the relationship between AQP4 and NGF in SCI, which may shed light on illustrating the role of AQP4 in SCI. These findings may also provide strategies of the clinical treatment for SCI in the future.


Subject(s)
Aquaporin 4/antagonists & inhibitors , Motor Activity/physiology , Nerve Growth Factor/metabolism , RNAi Therapeutics , Recovery of Function/physiology , Spinal Cord Injuries/therapy , Animals , Aquaporin 4/metabolism , Disease Models, Animal , Female , Genetic Vectors , Lentivirus/genetics , Random Allocation , Rats, Sprague-Dawley , Severity of Illness Index , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
20.
Sci Rep ; 5: 8486, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25686213

ABSTRACT

The recovery of motor function in rats is inhibited following contusion spinal cord injury (cSCI). However, the mechanism of tumour necrosis factor α (TNF-α) in motor function after cSCI associated with peroxiredoxin 6 (PRDX6) remains unknown. We randomly divided rats into four groups: sham, cSCI, vector and lentivirus mediating TNF-α RNA interference (TNF-α-RNAi-LV) group. The Basso, Beattie, Bresnahan (BBB) scale was used to evaluate motor function. Real-time quantitative PCR (qRT-PCR) and western blotting were used to detect the expression of TNF-α and PRDX6, which were located in neurons using immunohistochemistry (IHC) and immunofluorescence. Subsequently, lentiviral-mediated TNF-α was used to determine the role of TNF-αand the relationship of PRDX6 and TNF-α in cSCI. After cSCI, the motor capability of hind limbs disappeared and was followed by recovery of function. IHC analysis indicated that TNF-α and PRDX6 were primarily located in spinal cord neurons. TNF-α interference significantly improved neural behaviour and increased expression of PRDX6. Our study suggests that inhibition of TNF-α can promote the recovery of motor function. The underlying mechanism of TNF-α-promoted motor function may be connected with the up-regulation of PRDX6. This provides a new strategy or target for the clinical treatment of SCI in future.


Subject(s)
Motor Activity/genetics , Peroxiredoxin VI/genetics , RNA Interference , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Tumor Necrosis Factor-alpha/genetics , Animals , Blood-Brain Barrier/metabolism , Disease Models, Animal , Female , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Lentivirus/genetics , Peroxiredoxin VI/metabolism , Protein Transport , Rats , Spinal Cord Injuries/metabolism , Transduction, Genetic , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...