Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Mol Psychiatry ; 28(10): 4138-4150, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37188779

ABSTRACT

Depression is a mental illness frequently accompanied by disordered energy metabolism. A dysregulated hypothalamus pituitary adrenal axis response with aberrant glucocorticoids (GCs) release is often observed in patients with depression. However, the associated etiology between GCs and brain energy metabolism remains poorly understood. Here, using metabolomic analysis, we showed that the tricarboxylic acid (TCA) cycle was inhibited in chronic social defeat stress (CSDS)-exposed mice and patients with first-episode depression. Decreased mitochondrial oxidative phosphorylation was concomitant with the impairment of the TCA cycle. In parallel, the activity of pyruvate dehydrogenase (PDH), the gatekeeper of mitochondrial TCA flux, was suppressed, which is associated with the CSDS-induced neuronal pyruvate dehydrogenase kinase 2 (PDK2) expression and consequently enhanced PDH phosphorylation. Considering the well-acknowledged role of GCs in energy metabolism, we further demonstrated that glucocorticoid receptors (GR) stimulated PDK2 expression by directly binding to its promoter region. Meanwhile, silencing PDK2 abrogated glucocorticoid-induced PDH inhibition, restored the neuronal oxidative phosphorylation, and improved the flux of isotope-labeled carbon (U-13C] glucose) into the TCA cycle. Additionally, in vivo, pharmacological inhibition and neuron-specific silencing of GR or PDK2 restored CSDS-induced PDH phosphorylation and exerted antidepressant activities against chronic stress exposure. Taken together, our findings reveal a novel mechanism of depression manifestation, whereby elevated GCs levels regulate PDK2 transcription via GR, thereby impairing brain energy metabolism and contributing to the onset of this condition.


Subject(s)
Energy Metabolism , Pyruvate Dehydrogenase Complex , Humans , Mice , Animals , Pyruvate Dehydrogenase Complex/metabolism , Citric Acid Cycle/physiology , Brain/metabolism , Phosphorylation
2.
Amino Acids ; 54(1): 137-146, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34800175

ABSTRACT

Amino acids are one of the main metabolites in the body, and provide energy for the body and brain. The purpose of this study is to provide a profile of amino acid changes in the serum of patients with Moyamoya disease (MMD) and identify potential disease biomarkers. In this paper, we quantitatively determined the serum amino acid metabolic profiles of 43 MMD patients and 42 healthy controls (HCs). T test, multivariate statistical analysis, and receiver operating characteristic (ROC) curve analysis were used to identify candidate markers. Thirty-nine amino acids were quantified, and 12 amino acid levels differed significantly between the MMD patients and HCs. Moreover, based on ROC curve analysis, four amino acid (L-methionine, L-glutamic acid, ß-alanine and o-phosphoserine) biomarkers showed high sensitivity and specificity (AUC > 0.90), and showed the best sensitivity and specificity in MetaboAnalyst 5.0 using binary logistic regression analysis. We have provided serum amino acid metabolic profiles of MMD patients, and identified four potential biomarkers which may both provide clinicians with an objective diagnostic method for early detection of MMD and further our understanding of MMD pathogenesis.


Subject(s)
Moyamoya Disease , Amino Acids/metabolism , Biomarkers , Humans , Metabolome , Metabolomics/methods , ROC Curve
3.
Pharmacol Res ; 179: 106224, 2022 05.
Article in English | MEDLINE | ID: mdl-35430377

ABSTRACT

Ghrelin has recently been associated with the development of diabetes comorbid with depression, but its underlying molecular mechanisms remains poorly understood. Here, molecular and histological methods were applied both in vivo and in vitro studies to investigate the mechanisms of ghrelin in diabetes comorbid with depression. Our results demonstrated the anti-depressive, anxiolytic, and neuroprotective effects of ghrelin, as evidenced by the amelioration of anxiety- and depression-like behaviors, reduction in apoptosis, and preservation of neuron integrity in streptozotocin (STZ)-treated rats. STZ treatment induced M1-phenotypic microglial polarization, accompanied by neuroinflammation, which was reversed by ghrelin treatment. Further exploration showed that autophagy was inhibited, the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome and nuclear factor (NF)-κB signaling pathway were activated in STZ rats. In line with the in vivo results, ghrelin could suppress the NLRP3 inflammasome and NF-κB signaling pathway activation via the amelioration of impaired autophagic flux in microglial BV2 cells. Importantly, clinical evidence further verified the anti-inflammatory and antidepressant effects of ghrelin. Collectively, these results suggested that ghrelin ameliorates diabetes-associated behavioral deficits and NLRP3 inflammasome activation via autophagic flux enhancement, highlighting the importance of ghrelin as a potential target of immune regulation in diabetes comorbid with depression.


Subject(s)
Diabetes Mellitus , Inflammasomes , Animals , Autophagy , Ghrelin/pharmacology , Ghrelin/therapeutic use , Inflammasomes/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Streptozocin/pharmacology
4.
Mol Med ; 27(1): 118, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556021

ABSTRACT

BACKGROUND: The present study aimed to further explore the potential interaction between oxidative stress and autophagy in the progression of traumatic brain injury (TBI) and therapeutic mechanism of calcitriol, the active form of vitamin D (VitD). METHODS: Neuroprotective effects of calcitriol were examined following TBI. We further evaluated the impacts of TBI and calcitriol treatment on autophagic process and nuclear factor E2-related factor 2 (Nrf2) signaling. RESULTS: We found that treatment of calcitriol markedly ameliorated the neurological deficits and histopathological changes following TBI. The brain damage impaired autophagic flux and impeded Nrf2 signaling, the major regulator in antioxidant response, consequently leading to uncontrolled and excessive oxidative stress. Meanwhile, calcitriol promoted autophagic process and activated Nrf2 signaling as evidenced by the reduced Keap1 expression and enhanced Nrf2 translocation, thereby mitigating TBI-induced oxidative damage. In support, we further found that chloroquine (CQ) treatment abrogated calcitriol-induced autophagy and compromised Nrf2 activation with increased Keap1 accumulation and reduced expression of Nrf2-targeted genes. Additionally, both CQ treatment and Nrf2 genetic knockout abolished the protective effects of calcitriol against both TBI-induced neurological deficits and neuronal apoptosis. CONCLUSIONS: Therefore, our work demonstrated a neuroprotective role of calcitriol in TBI by triggering Nrf2 activation, which might be mediated by autophagy.


Subject(s)
Autophagy/drug effects , Brain Injuries, Traumatic/prevention & control , Calcitriol/pharmacology , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Disease Models, Animal , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Male , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/prevention & control , Mice, Knockout , Microscopy, Electron, Transmission , NF-E2-Related Factor 2/genetics , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/prevention & control , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Signal Transduction/genetics , Vitamins/pharmacology
5.
Mediators Inflamm ; 2020: 5847478, 2020.
Article in English | MEDLINE | ID: mdl-32410857

ABSTRACT

Systemic inflammation has been shown to play a pivotal role in the pathogenesis of moyamoya disease (MMD). Brain-gut peptides exhibit regulatory effects in the secretion of proinflammatory cytokines. To investigate the association between brain-gut peptides and inflammation in the occurrence of MMD, 41 patients with MMD, as well as 74 age- and sex-matched healthy individuals were enrolled. The levels of four brain-gut peptides (vasoactive intestinal polypeptide (VIP), cholecystokinin (CCK), somatostatin (SST), substance P (SP)) and three proinflammatory cytokines (interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), IL-12) in the serum and cerebrospinal fluid (CSF) were measured using the enzyme-linked immunosorbent assay. The associations between brain-gut peptides and proinflammatory cytokines were estimated according to the multiple linear regression and correlation analyses. MMD patients exhibited significantly lower levels of VIP, CCK, and SST and higher levels of IL-1ß, TNF-α, and IL-12 in the serum compared with healthy controls. Multiple logistic regression analysis showed that decreased VIP, CCK, and SST levels were independent predictors of the occurrence of MMD. Negative correlations were observed between the VIP and proinflammatory cytokines, including IL-1ß, TNF-α, and IL-12 (serum vs. CSF). Significant negative correlations were also found between CCK and IL-1ß, as well as IL-12 (serum vs. CSF). SST was negatively correlated with IL-1ß and TNF-α in the serum and IL-1ß only in the CSF. In addition, the levels of VIP, CCK, SST, and proinflammatory cytokines IL-1ß and TNF-α in the serum were correlated with those measured in the CSF. Collectively, lower levels of VIP, CCK, and SST may be associated with the pathogenesis of MMD and act as clinically useful biomarkers along with the levels of proinflammatory cytokines.


Subject(s)
Brain/metabolism , Cytokines/metabolism , Gastrointestinal Microbiome , Intestines/pathology , Moyamoya Disease/metabolism , Peptides/metabolism , Adult , Biomarkers/blood , Case-Control Studies , Female , Humans , Inflammation , Male , Middle Aged , ROC Curve , Regression Analysis , Tumor Necrosis Factor-alpha/metabolism , Vasoactive Intestinal Peptide
6.
Cell Physiol Biochem ; 42(1): 137-144, 2017.
Article in English | MEDLINE | ID: mdl-28505619

ABSTRACT

BACKGROUND/AIMS: Intracerebral hemorrhage (ICH) occurs in hypertensive patients and results in high rates of mortality and disability. This study determined whether bone marrow mesenchymal stem cell (BMSC) transplantation affects axonal regeneration and examined the underlying mechanisms after the administration of PD98059 (p-ERK1/2 inhibitor) or/ and LY294002 (PI3K inhibitor). The hypothesis that was intended to be tested was that BMSC transplantation regulates the expression of growth-associated protein-43 (GAP-43) via the ERK1/2 and PI3K/Akt signaling pathways. METHODS: Seventy-five male rats (250-280 g) were subjected to intracerebral blood injection and then randomly received a vehicle, BMSCs, PD98059 or LY294002 treatment. Neurological deficits were evaluated prior to injury and at 1, 3 and 7 days post-injury. The expression of GAP-43, Akt, p-Akt, ERK1/2, and p-ERK1/2 proteins was measured by western blot analysis. RESULTS: BMSC transplantation attenuated neurological deficits 3-7 days post-ICH. The expression of GAP-43 was increased 3 days following BMSC transplantation. However, this increase was inhibited by either PD98059 or LY294002 treatment. Treatment with both PD98059 and LY294002 was more effective than was treatment with an individual compound. CONCLUSION: BMSC transplantation could attenuate neurological deficits and activate axonal regeneration in this rat ICH model. The protective effects might be associated with increased GAP-43 expression by activating both the ERK1/2 and PI3K/Akt signaling pathways.


Subject(s)
Cerebral Hemorrhage/therapy , GAP-43 Protein/metabolism , Mesenchymal Stem Cell Transplantation , Signal Transduction , Animals , Axons/physiology , Bone Marrow Cells/cytology , Cells, Cultured , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/pathology , Chromones/pharmacology , Disease Models, Animal , Flavonoids/pharmacology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Regeneration , Signal Transduction/drug effects , Up-Regulation/drug effects
7.
Cell Physiol Biochem ; 42(5): 1888-1896, 2017.
Article in English | MEDLINE | ID: mdl-28772270

ABSTRACT

BACKGROUND/AIMS: Traumatic brain injury (TBI) is a major public health problem in the world and causes high rates of mortality and disability. Recent evidence suggests that vitamin D (VD) has neuroprotective actions and can promote function recovery after TBI. In vitro and in vivo studies have demonstrated that autophagy could be enhanced following supplementation with an active metabolite of VD (calcitriol). However, it is unclear whether autophagy participates in the protective effects of calcitriol after TBI. To test this hypothesis, we examined the protective effects of calcitriol on TBI-induced neurological impairment and further investigated whether calcitriol could modulate autophagy dysfunction-mediated cell death in the cortex region of rat brain. METHODS: Eighty-five male rats (250-280 g) were randomly assigned to sham (n=15), TBI model (TBI, n=35) and calcitriol treatment (calcitriol, n=35) groups. Rats were injected intraperitoneally with calcitriol (1 µg/kg) at 30 min, 24 h and 48 h post-TBI in the calcitriol group. The lysosomal inhibitor, chloroquine (CQ), was used to evaluate autophagic flux in the TBI and calcitriol groups. Neurological functions were evaluated via the modified neurological severity score test at 1-7 days after TBI or sham operation, and the terminal deoxynucleotidyl transferase-mediated FITC-dUTP nick-end labeling method was used to evaluate the ability of calcitriol to inhibit apoptosis. The expression of VDR, LC3 and p62 proteins was measured by western blot analysis at 1, 3 and 7 days post-injury Results: Calcitriol treatment attenuated mNSS at 2-7 days post-TBI (P < 0.05 versus TBI group). Calcitriol dramatically increased VDR protein expression compared with the untreated counterparts at 1, 3 and 7 days post-TBI (P < 0.05). The rate of apoptotic cells in calcitriol-treated rats was significantly reduced compared to that observed in the TBI group (P < 0.05). The LC3II/LC3I ratio was decreased in the cortex region at 1, 3 and 7 days post-TBI in rats treated with calcitriol (p < 0.05 versus TBI group), and the p62 expression was also attenuated (p < 0.05 versus TBI group). The LC3II/LC3I ratio in the calcitriol group was significantly increased when pretreated with CQ (P < 0.05). CONCLUSION: Calcitriol treatment activated VDR protein expression and attenuated neurological deficits in this rat TBI model. The protective effects might be associated with the restoration of autophagy flux and the decrease in apoptosis in the cortex region of rat brain.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Brain Injuries, Traumatic/pathology , Calcitriol/pharmacology , Receptors, Calcitriol/metabolism , Animals , Brain Injuries, Traumatic/metabolism , Chloroquine/toxicity , Disease Models, Animal , Injections, Intraperitoneal , Male , Membrane Glycoproteins/metabolism , Microtubule-Associated Proteins/metabolism , Neuroprotective Agents/pharmacology , Nuclear Pore Complex Proteins/metabolism , Rats , Receptors, Calcitriol/agonists
8.
Cell Mol Neurobiol ; 37(1): 155-168, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26961544

ABSTRACT

Accumulating evidence indicates that autophagy and inflammatory responses contributes to secondary brain injury after traumatic brain injury (TBI), and toll-like receptor 4 (TLR4) is considered to involvement of this cascade and plays an important role. The present study was designed to determine the hypothesis that administration of resatorvid (TAK-242), a TLR4 antagonist, might provide a neuroprotective effect by inhibit TLR4-mediated pathway in a TBI rat model. Rat subjected to controlled cortical impact injury were injected with TAK-242 (0.5 mg/kg, i.v. injected) 10 min prior to injury. The results demonstrated that TAK-242 treatment significantly attenuated TBI-induced neurons loss, brain edema, and neurobehavioral impairment in rats. Immunoblotting analysis showed that TAK-242 treatment reduced TBI-induced TLR4, Beclin 1, and LC3-II levels, and maintained p62 levels at 24 h. Double immunolabeling demonstrated that LC3 dots co-localized with the hippocampus pyramidal neurons, and TLR4 was localized with the hippocampus neurons and astrocytes. In addition, the expression of TLR4 downstream signaling molecules, including MyD88, TRIF, NF-κB, TNF-α, and IL-1ß, was significantly downregulated in hippocampus tissue by Western blot analysis. In conclusion, our findings indicate that pre-injury treatment with TAK-242 could inhibit neuronal autophagy and neuroinflammation responses in the hippocampus in a rat model of TBI. The neuroprotective effects of TAK-242 may be related to modulation of the TLR4-MyD88/TRIF-NF-κB signaling pathway. Furthermore, the study also suggests that TAK-242, an attractive potential drug, may be a promising drug candidate for TBI.


Subject(s)
Autophagy/physiology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/prevention & control , Neuroprotective Agents/therapeutic use , Sulfonamides/therapeutic use , Toll-Like Receptor 4/biosynthesis , Animals , Autophagy/drug effects , Brain Injuries, Traumatic/pathology , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/pharmacology , Treatment Outcome
9.
Neurochem Res ; 42(11): 3296-3309, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28786047

ABSTRACT

Neuronal autophagy and inflammatory responses are important in the pathogenesis of traumatic brain injury (TBI), and toll-like receptor 4 (TLR4) may play an important role in the related molecular cascade. The present study investigated the protective effect of apocynin, an inhibitor of NADPH oxidase, in a TBI rat model and further examined neuronal autophagy and the TLR4-mediated pathway. Adult male Sprague-Dawley rats were subjected to controlled cortical impact injury and intraperitoneally injected with apocynin (50 mg/kg) immediately after the trauma. In addition to motor and behavioral studies, brain water content and histology analyses were performed. Expression of autophagy-related proteins as well as TLR4/NF-κB signaling and inflammatory mediators was analyzed. The apocynin treatment significantly attenuated TBI-induced motor and behavioral impairment, brain edema and neuronal damage in rats. Immunohistochemical and Western blot analyses revealed that apocynin treatment significantly reduced the expression of NOX2, LC3 and Beclin1 in the hippocampus at 12-48 h after injury. Double immunolabeling demonstrated that apocynin decreased the co-localization of LC3 or TLR4-positive cells with hippocampal neurons at 24 h following TBI. In addition, CD11b (microglial marker) and GFAP (astrocyte marker)-immunopositive cells were also clearly decreased in hippocampal tissues. Meanwhile, protein levels of TLR4, NF-κB p65, TNF-α and IL-1ß were found to be significantly downregulated by Western blot analysis. In conclusion, our findings indicate that the protective effects of apocynin may be related to modulation of neuronal autophagy and the TLR4/NF-κB signaling pathway.


Subject(s)
Acetophenones/therapeutic use , Autophagy/physiology , Brain Injuries, Traumatic/metabolism , NF-kappa B/metabolism , Neurons/metabolism , Toll-Like Receptor 4/metabolism , Acetophenones/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Autophagy/drug effects , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Male , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Rotarod Performance Test/methods , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Neurol Sci ; 35(5): 695-700, 2014 May.
Article in English | MEDLINE | ID: mdl-24277205

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of mortality and disability in children and young adults worldwide. Neurologic impairment is caused by both immediate brain tissue disruption and post-injury cellular and molecular events that worsen the primary neurologic insult. The ß-lactam antibiotic ceftriaxone (CTX) has been reported to induce neuroprotection in animal models of diverse neurologic diseases via up-regulation of GLT-1. However, no studies have addressed the neuroprotective role of CTX in the setting of TBI, and whether the mechanism is involved in the modulation of neuronal autophagy remains totally unclear. The present study was designed to determine the hypothesis that administration of CTX could significantly enhance functional recovery in a rat model of TBI and whether CTX treatment could up-regulate GLT-1 expression and suppress post-TBI neuronal autophagy. The results demonstrated that daily treatment with CTX attenuated TBI-induced brain edema and cognitive function deficits in rats. GLT-1 is down-regulated following TBI and this phenomenon can be reversed by treatment of CTX. In addition, we also found that CTX significantly reduced autophagy marker protein, LC3 II, in hippocampus compared to the TBI group. These results suggest that CTX might provide a new therapeutic strategy for TBI and this protection might be associated with up-regulation of GLT-1 and suppression of neuronal autophagy.


Subject(s)
Brain Injuries/drug therapy , Ceftriaxone/pharmacology , Neuroprotective Agents/pharmacology , Animals , Autophagy/drug effects , Autophagy/physiology , Brain Edema/drug therapy , Brain Edema/etiology , Brain Edema/pathology , Brain Edema/physiopathology , Brain Injuries/complications , Brain Injuries/pathology , Brain Injuries/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Disease Models, Animal , Excitatory Amino Acid Transporter 2/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Male , Maze Learning/drug effects , Maze Learning/physiology , Microtubule-Associated Proteins/metabolism , Random Allocation , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Up-Regulation/drug effects
11.
J Res Med Sci ; 19(4): 304-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-25097601

ABSTRACT

BACKGROUND: Despite increasing acceptance of endovascular coiling for treating anterior communicating artery (ACoA) aneurysms, anterior circulation cerebral infarction (ACI) after embolization remains a limitation. With higher incidence, higher morbidity and higher mortality, it is one of the main factors influencing the ACoA aneurysms prognosis. Determining the risk factors leading to ACI after embolization will have clinical significance. Through retrospective case analysis, this study investigated the risk factors related to ACI after embolization in order to provide information to serve the clinical practice. MATERIALS AND METHODS: A retrospective review was performed of patients who had undergone coiling of ACoA aneurysms from 2008 to 2012. All patients had ruptured prior to the completion of embolization. Cases with acute stroke symptoms without alternative diagnoses after embolization were diagnosed as ACI. A total of 32 risk factors such as age, sex, hypertension, diabetes mellitus, modified Fisher grade, Hunt-Hess grade, ventricular hemorrhage, etc. were analyzed using univariate and logistic regression analysis. RESULTS: Univariate analysis showed that negative fluid volume balance (P = 0.041 <0.05) and modified Fisher grade (P = 0.049 <0.05) reached statistical significance, suggesting that they might be risk factors for ACI after embolization. Multiple logistic regression analysis showed that modified Fisher grade was significantly associated with ACI after embolization, suggesting that it was an independent risk factor (odds ratios (OR): 4.968, 95% confidence intervals (CI): 1.013-24.360, P = 0.048). CONCLUSION: Modified Fisher grade is an independent risk factor for ACI after embolization.

12.
Heliyon ; 10(11): e32022, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38868045

ABSTRACT

Moyamoya disease currently lacks a suitable method for early clinical screening.This study aimed to identify a simple and feasible clinical screening index by investigating microRNAs carried by peripheral blood exosomes. Experimental subjects participated in venous blood collection, and exosomes were isolated using Exquick-related technology. Sequencing was performed on the extracted exosomal ribonucleic acids (RNAs) to identify differential microRNAs. Verification of the results involved selecting relevant samples from the genetic database. The study successfully pinpointed a potential marker for early screening, hsa-miR-328-3p + hsa-miR-200c-3p carried by peripheral blood exosomes. Enrichment analysis of target genes revealed associations with intercellular junctions, impaired cytoskeletal regulation, and increased fibroblast proliferation, leading to bilateral internal carotid artery neointimal expansion and progressive stenosis. These findings establish the diagnostic value of hsa-miR-328-3p+hsa-miR-200c-3p in screening moyamoya disease, while also contributing to a deeper understanding of its underlying pathophysiology. Significant differences in microRNA expressions derived from peripheral blood exosomes were observed between moyamoya disease patients and control subjects. Consequently, the utilization of peripheral blood exosomes, specifically hsa-miR-328-3p + hsa-miR-200c-3p, holds potential for diagnostic screening purposes.

13.
Drug Des Devel Ther ; 18: 2435-2447, 2024.
Article in English | MEDLINE | ID: mdl-38915864

ABSTRACT

Purpose: SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of irinotecan, has been extensively studied in drug delivery systems. However, its impact on neural metabolism remains unclear. This study aims to investigate the toxic effects of SN-38 on mouse brain metabolism. Methods: Male mice were divided into an SN-38 group and a control group. The SN-38 group received SN-38 (20 mg/kg/day) via intraperitoneal injection, while the control group was given an equal volume of a blank solvent mixture (DMSO and saline, ratio 1:9). Gas chromatography-mass spectrometry (GC-MS) was employed to analyze differential metabolites in the cortical and hippocampal regions of the SN-38-treated mice. Results: SN-38 induced metabolic disturbances in the central nervous system. Eighteen differential metabolites were identified in the hippocampus and twenty-four in the cortex, with six common to both regions. KEGG pathway enrichment analysis revealed statistically significant alterations in six metabolic pathways in the hippocampus and ten in the cortex (P<0.05). Conclusion: This study is the first to demonstrate the neurotoxicity of SN-38 in male mice through metabolomics. Differential metabolites in the hippocampal and cortical regions were closely linked to purine metabolism, pyrimidine metabolism, amino acid metabolism, and glyceride metabolism, indicating disruptions in the blood-brain barrier, energy metabolism, and central signaling pathways.


Subject(s)
Brain , Irinotecan , Metabolomics , Animals , Male , Irinotecan/pharmacology , Mice , Brain/metabolism , Brain/drug effects , Gas Chromatography-Mass Spectrometry , Injections, Intraperitoneal
14.
Psychopharmacology (Berl) ; 241(7): 1329-1343, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38411637

ABSTRACT

RATIONALE: Microglia-mediated neuroinflammation is a vital hallmark in progression of depression, while calcitriol exerts anti-inflammatory effects in the brain. The activation of the P2X7 receptor has an important link to neuroinflammation. However, it is unclear whether calcitriol treatment exerts anti-inflammatory effects in association with P2X7R activation. OBJECTIVE: In this study, we assessed the antidepressive and neuroprotective effects of calcitriol on lipopolysaccharide (LPS)-mediated depressive-like behavior, neuroinflammation, and neuronal damage. METHODS: In in vitro experiments, the BV2 cells were exposed to LPS, and the protective effects of calcitriol were assessed. For in vivo experiment, thirty-two male C57BL/6 mice were divided into four groups of control, calcitriol, LPS and LPS + calcitriol. Calcitriol was administered at 1 µg/kg for 14 days and LPS at 1 mg/kg once every other day for 14 days. The control group mice were given equal volumes of vehicles. All treatments were delivered intraperitoneally. RESULTS: The in vitro experiments showed calcitriol inhibited the release of inflammatory mediators induced by LPS in BV2 cells. The in vivo experiments revealed that calcitriol alleviated LPS-induced behavioral abnormalities and spatial learning impairments. Moreover, calcitriol treatment reduced the mRNA levels of pro-inflammatory cytokines, while increasing anti-inflammatory cytokine levels in the hippocampus. Our results further revealed that calcitriol administration attenuated LPS-induced microglia activation by suppressing P2X7R/NLRP3/caspase-1 signaling. Moreover, calcitriol inhibited apoptosis of neurons in the hippocampus as evidenced by expression of apoptosis-related proteins and TUNEL assay. CONCLUSIONS: Collectively, our findings demonstrated that calcitriol exerts antidepressive and neuroprotective effects through the suppression of the P2X7R/NLRP3/caspase-1 pathway both in LPS-induced inflammation models in vitro and in vivo.


Subject(s)
Calcitriol , Caspase 1 , Depression , Lipopolysaccharides , Mice, Inbred C57BL , Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Receptors, Purinergic P2X7 , Animals , Receptors, Purinergic P2X7/metabolism , Calcitriol/pharmacology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Depression/drug therapy , Depression/metabolism , Depression/chemically induced , Caspase 1/metabolism , Microglia/drug effects , Microglia/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Signal Transduction/drug effects , Neuroprotective Agents/pharmacology , Behavior, Animal/drug effects , Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Cell Line , Disease Models, Animal
15.
MedComm (2020) ; 4(6): e410, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37916034

ABSTRACT

Exosomes, membrane-enclosed vesicles, are secreted by all types of cells. Exosomes can transport various molecules, including proteins, lipids, functional mRNAs, and microRNAs, and can be circulated to various recipient cells, leading to the production of local paracrine or distal systemic effects. Numerous studies have proved that exosomes can pass through the blood-brain barrier, thus, enabling the transfer of peripheral substances into the central nervous system (CNS). Consequently, exosomes may be a vital factor in the exchange of information between the periphery and CNS. This review will discuss the structure, biogenesis, and functional characterization of exosomes and summarize the role of peripheral exosomes deriving from tissues like the lung, gut, skeletal muscle, and various stem cell types in communicating with the CNS and influencing the brain's function. Then, we further discuss the potential therapeutic effects of exosomes in brain diseases and the clinical opportunities and challenges. Gaining a clearer insight into the communication between the CNS and the external areas of the body will help us to ascertain the role of the peripheral elements in the maintenance of brain health and illness and will facilitate the design of minimally invasive techniques for diagnosing and treating brain diseases.

16.
J Toxicol ; 2023: 5660481, 2023.
Article in English | MEDLINE | ID: mdl-37575636

ABSTRACT

Objective: It is well known that paclitaxel (PTX)-induced neurotoxicity seriously affects the quality of life of patients and is the main reason for reducing the dose of chemotherapy or even stopping chemotherapy. The current data are limited, and further information is required for practice and verification. The aims of this study were to clarify the molecular mechanism underlying PTX-induced neurotoxicity by combining in vivo and in vitro metabolomics studies and provide new targets for the prevention and treatment of PTX-induced neurotoxicity. Methods: In the in vivo study, a PTX-induced neurotoxicity mouse model was established by intraperitoneal injection of PTX (6 mg/kg every three days) for two consecutive weeks. After verification by water maze tests and HE staining of pathological sections, hippocampal metabolites were measured and the differential metabolites and related metabolic pathways were identified by multivariate statistical analysis. In the in vitro study, we investigated the effects of PTX on mouse hippocampal neuron cells, assessing the concentration and time of administration by MTT assays. After modeling, the relevant metabolites in the TCA cycle were quantified by targeted metabolomics using stable isotope labeling. Finally, the key enzymes of the TCA cycle in tissues and cells were verified by RT-PCR. Results: Administration of PTX to model mice resulted in neurological damage, shown by both water-maze tests and hippocampal tissue sections. Twenty-four metabolites and five associated metabolic pathways were found to differ significantly between the hippocampal tissues of the model and control groups. These included metabolites and pathways related to the TCA cycle and pyruvate metabolism. Metabolomics analysis using stable isotope labeling showed significant changes in metabolites associated with the TCA cycle compared with the control group (P < 0.05). Finally, RT-PCR verified that the expression of key enzymes in the TCA cycle was changed to different degrees in both hippocampal tissues and cells. Conclusion: Our results showed that PTX neurotoxicity in hippocampal tissue and neuron cells was associated with inhibition of the TCA cycle. This inhibition leads to brain insufficiency and impaired metabolism, resulting in various neurotoxic symptoms.

17.
Cell Death Dis ; 14(9): 643, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37773169

ABSTRACT

The interaction between cerebral endothelial cells (CEC) and brain parenchymal cells is critical to maintain neurovascular homeostasis, whereas extracellular vesicles (EVs) are essential to mediate the cell-cell communication. Previous researches demonstrated that CEC-derived EVs (CEC-EVs) confer neuroprotective actions. However, the molecular mechanisms remain unknown. In this study, we isolated EVs from CEC and assessed their immune-regulatory actions in microglial cells and mice following lipopolysaccharide (LPS) exposure. We found that CEC-EVs treatment significantly ameliorated LPS-induced inflammatory activation, shifting microglial polarization from pro-inflammatory phenotype to anti-inflammatory phenotype. Meanwhile, microglial cells can effectively internalize CEC-EVs and this process was further enhanced by immune activation. Next, the miRNA microarray analysis revealed that CEC-EVs increased expression of miR-672-5p, which was demonstrated to be the cargo of CEC-EVs. TGFß-activated kinase 1 (TAK1)-binding proteins 2 (TAB2) was identified to be the target of miR-672-5p. Through inhibiting TAB2, miR-672-5p derived from CEC-EVs suppressed TAK1-TAB signaling and thereby mitigating the downstream NF-κB activation. Furthermore, we found that by delivering miR-672-5p, CEC-EVs promoted autophagy and hence stimulating autophagic degradation of NLRP3 inflammasome. Our work firstly revealed the neuroimmune-modulating actions of CEC-EVs and further demonstrated that miR-672-5p secreted from CEC-EVs inhibits microglial pro-inflammatory polarization and facilitates autophagic process via targeting TAB2.


Subject(s)
Extracellular Vesicles , MicroRNAs , Animals , Mice , Endothelial Cells , Lipopolysaccharides/pharmacology , Microglia , Autophagy , MicroRNAs/genetics
18.
Turk Neurosurg ; 32(3): 347-356, 2022.
Article in English | MEDLINE | ID: mdl-34936071

ABSTRACT

AIM: To review the correlation between vitamin D (VD) and several common neurological disorders with the aim of providing directions and ideas for using VD to treat neurological disorders. MATERIAL AND METHODS: VD, 1,25-dihydroxyvitamin D3, stroke, epilepsy, and cognitive dysfunction were used as keywords. The PubMed and Embase databases were searched for articles published from 2010 to 2021. The inclusion criteria were as follows: clear introduction of the research sample, detailed explanation of the sample selection in the research, intervention, and control measures, and available odds ratio and 95% confidence interval. The exclusion criteria were as follows: duplicate reports, defects in research design and poor quality, incomplete data and unclear results, and unmodifiable errors in the statistical method. RESULTS: Initially, 1,360 articles were retrieved from the PubMed and Embase databases. Finally, 81 articles were included, 76 of which were published within the last 5 years. CONCLUSION: VD deficiency is very common in the population and is associated with a variety of neurological diseases. VD, a neuroactive steroid, plays an important role in the protection of the central nervous system. In contrast, stroke can cause epilepsy and varying degrees of changes in cognitive function. Furthermore, seizure and epilepsy can cause changes in cognitive function. The degree of alteration in cognitive function affects the occurrence and progression of stroke and epilepsy. Therefore, VD can be used for the comprehensive treatment of neurological diseases.


Subject(s)
Epilepsy , Stroke , Cognition , Humans , Seizures , Stroke/complications , Vitamin D/therapeutic use
19.
Tissue Cell ; 76: 101772, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35272241

ABSTRACT

Extracellular vesicles (EVs) are biologically active membrane vesicles secreted by many cells in the body. A variety of nucleic acids, proteins, and other biologically active substances in EVs can be used to exchange and transmit information between cells, thereby affecting the progression of various diseases. Numerous studies have demonstrated that EVs not only regulate changes in brain physiology but also regulate synaptic plasticity and neuronal regeneration in traumatic brain injury (TBI), which opens a new approach for the treatment of TBI. In view of the fact that most human cells can secrete EVs, and relevant experiments have proved that different doses of EVs have different therapeutic effects on TBI. To this end, this paper reviews the therapeutic effects of EVs from different cell sources and their doses on TBI.


Subject(s)
Brain Injuries, Traumatic , Extracellular Vesicles , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Extracellular Vesicles/metabolism , Humans , Neurons
20.
Drug Des Devel Ther ; 16: 1915-1930, 2022.
Article in English | MEDLINE | ID: mdl-35747443

ABSTRACT

Purpose: Sodium valproate (VPA) is the most widely used broad-spectrum antiepileptic first-line drug in clinical practice and is effective against various types of epilepsy. However, VPA can induce severe cardiotoxicity, nephrotoxicity, hepatotoxicity, and neurotoxicity, which limits its use. Metabolomic studies of VPA-induced toxicity have focused primarily on changes in serum and urine metabolites but have not evaluated changes in major organs or tissues. Methods: Central target tissues (intestine, lung, liver, hippocampus, cerebral cortex, inner ear, spleen, kidney, heart, and serum) were analyzed using gas chromatography mass spectrometry to comprehensively evaluate VPA toxicity in mouse models. Results: Multivariate analyses, including orthogonal projections of the latent structure and Student's t test, indicated that depending on the matrix used in the study (the intestine, lung, liver, hippocampus, cerebral cortex, inner ear, spleen, kidney, heart or serum) the number of metabolites differed, the lung being the poorest and the kidney the richest in number. Conclusion: These metabolites were closely related and were found to participate in 12 key pathways related to amino acid, fatty acid, and energy metabolism, revealing that the toxic mechanism of VPA may involve oxidative stress, inflammation, amino acid metabolism, lipid metabolism, and energy disorder.


Subject(s)
Chemical and Drug Induced Liver Injury , Valproic Acid , Amino Acids , Animals , Anticonvulsants/therapeutic use , Anticonvulsants/toxicity , Chemical and Drug Induced Liver Injury/drug therapy , Gas Chromatography-Mass Spectrometry/methods , Humans , Male , Metabolomics/methods , Mice , Valproic Acid/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL