Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Brain Sci ; 13(2)2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36831824

ABSTRACT

BACKGROUND: Several complex cellular and gene regulatory processes are involved in peripheral nerve repair. This study uses bioinformatics to analyze the differentially expressed genes (DEGs) in the satellite glial cells of mice following sciatic nerve injury. METHODS: R software screens differentially expressed genes, and the WebGestalt functional enrichment analysis tool conducts Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomics (KEGG) pathway analysis. The Search Tool for the Retrieval of Interacting Genes/Proteins constructs protein interaction networks, and the cytoHubba plug-in in the Cytoscape software predicts core genes. Subsequently, the sciatic nerve injury model of mice was established and the dorsal root ganglion satellite glial cells were isolated and cultured. Satellite glial cells-related markers were verified by immunofluorescence staining. Real-time polymerase chain reaction assay and Western blotting assay were used to detect the mRNA and protein expression of Sox9 in satellite glial cells. RESULTS: A total of 991 DEGs were screened, of which 383 were upregulated, and 508 were downregulated. The GO analysis revealed the processes of biosynthesis, negative regulation of cell development, PDZ domain binding, and other biological processes were enriched in DEGs. According to the KEGG pathway analysis, DEGs are primarily involved in steroid biosynthesis, hedgehog signaling pathway, terpenoid backbone biosynthesis, American lateral skeleton, and melanoma pathways. According to various cytoHubba algorithms, the common core genes in the protein-protein interaction network are Atf3, Mmp2, and Sox9. Among these, Sox9 was reported to be involved in the central nervous system and the generation and development of astrocytes and could mediate the transformation between neurogenic and glial cells. The experimental results showed that satellite glial cell marker GS were co-labeled with Sox9; stem cell characteristic markers Nestin and p75NTR were labeled satellite glial cells. The mRNA and protein expression of Sox9 in satellite glial cells were increased after sciatic nerve injury. CONCLUSIONS: In this study, bioinformatics was used to analyze the DEGs of satellite glial cells after sciatic nerve injury, and transcription factors related to satellite glial cells were screened, among which Sox9 may be associated with the fate of satellite glial cells.

2.
Bioengineered ; 13(6): 14799-14814, 2022 06.
Article in English | MEDLINE | ID: mdl-36420646

ABSTRACT

Stroke is a main cause of disability and death among adults in China, and acute ischemic stroke accounts for 80% of cases. The key to ischemic stroke treatment is to recanalize the blocked blood vessels. However, more than 90% of patients cannot receive effective treatment within an appropriate time, and delayed recanalization of blood vessels causes reperfusion injury. Recent research has revealed that ischemic postconditioning has a neuroprotective effect on the brain, but the mechanism has not been fully clarified. Long non-coding RNAs (lncRNAs) have previously been associated with ischemic reperfusion injury in ischemic stroke. LncRNAs regulate important cellular and molecular events through a variety of mechanisms, but a comprehensive analysis of potential lncRNAs involved in the brain protection produced by ischemic postconditioning has not been conducted. In this review, we summarize the common mechanisms of cerebral injury in ischemic stroke and the effect of ischemic postconditioning, and we describe the potential mechanisms of some lncRNAs associated with ischemic stroke.


Subject(s)
Brain Injuries , Ischemic Postconditioning , Ischemic Stroke , RNA, Long Noncoding , Reperfusion Injury , Adult , Humans , RNA, Long Noncoding/genetics
3.
Biosci Rep ; 42(9)2022 09 30.
Article in English | MEDLINE | ID: mdl-35993194

ABSTRACT

Embolic stroke (ES) is characterized by high morbidity and mortality. Its mortality predictors remain unclear. The present study aimed to use machine learning (ML) to identify the key predictors of mortality for ES patients in the intensive care unit (ICU). Data were extracted from two large ICU databases: Medical Information Mart for Intensive Care (MIMIC)-IV for training and internal validation, and eICU Collaborative Research Database (eICU-CRD) for external validation. We developed predictive models of ES mortality based on 15 ML algorithms. We relied on the synthetic minority oversampling technique (SMOTE) to address class imbalance. Our main performance metric was area under the receiver operating characteristic (AUROC). We adopted recursive feature elimination (RFE) for feature selection. We assessed model performance using three disease-severity scoring systems as benchmarks. Of the 1566 and 207 ES patients enrolled in the two databases, there were 173 (15.70%), 73 (15.57%), and 36 (17.39%) hospital mortality in the training, internal validation, and external validation cohort, respectively. The random forest (RF) model had the largest AUROC (0.806) in the internal validation phase and was chosen as the best model. The AUROC of the RF compact (RF-COM) model containing the top six features identified by RFE was 0.795. In the external validation phase, the AUROC of the RF model was 0.838, and the RF-COM model was 0.830, outperforming other models. Our findings suggest that the RF model was the best model and the top six predictors of ES hospital mortality were Glasgow Coma Scale, white blood cell, blood urea nitrogen, bicarbonate, age, and mechanical ventilation.


Subject(s)
Critical Illness , Embolic Stroke , Hospital Mortality , Humans , Intensive Care Units , Machine Learning
4.
Brain Res Bull ; 185: 39-48, 2022 07.
Article in English | MEDLINE | ID: mdl-35452749

ABSTRACT

Remote ischemic postconditioning (RIPostC) is a protective procedure for brain damage caused by ischemia/reperfusion (IR), yet the mechanism of this treatment remains to be elucidated. Circular RNAs (circRNAs) are endogenous non-coding RNAs that have recently been recognized to play vital roles in ischemic brain injury. The aim of this study was to explore the role of circRNAs in the protective mechanism of RIPostC and to analyze the circRNA-microRNA (miRNA) regulation network in RIPostC. Nine rats were assigned randomly into three groups (three rats per group): sham, IR, and RIPostC. Their brain tissues were extracted for next-generation RNA sequencing and bioinformatics analysis was performed for two comparisons: sham vs. IR and IR vs. RIPostC. The expression patterns of selected circRNAs and miRNAs were validated by quantitative real-time PCR (qPCR). We detected 82 upregulated and 51 downregulated circRNAs and 137 upregulated and 127 downregulated miRNAs in the IR group compared with the sham group, and 41 upregulated and 100 downregulated circRNAs and 45 upregulated and 64 downregulated miRNAs in the RIPostC group compared with the IR group. The proposed competitive endogenous RNA (ceRNA) network, which included 24 circRNAs, 20 miRNAs, and 145 mRNAs, indicated that the dysregulated circRNAs played important roles in brain IR injury. On the basis of the expression patterns of selected circRNAs, miRNAs, and mRNAs obtained by qPCR, we proposed a circRNA_0002286-miR-124-3p-VLCAD pathway. In PC12 cell, the expression level of miR-124-3p was significantly upregulated when the expression of circRNA_0002286 was repressed and the expression level of VLCAD (very-long chain acyl-CoA dehydrogenase) was significantly downregulated, which suggested that circRNA_0002286 may act as a miRNA sponge for miR-124-3p to regulate the expression of VLCAD. We found that upregulation of circRNA_0002286 attenuated IR injury and was associated with downregulation of miR-124-3p and upregulation of VLCAD. This is the first time that circRNAs have been shown to be closely related to brain IR injury and RIPostC and suggests that targeting the circRNA_0002286-miR-124-3p-VLCAD pathway might attenuate brain IR injury.


Subject(s)
Brain Ischemia , Ischemic Postconditioning , MicroRNAs , Reperfusion Injury , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Brain/metabolism , Brain Ischemia/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Messenger/metabolism , Rats , Reperfusion Injury/genetics
5.
Neural Regen Res ; 17(6): 1299-1309, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34782575

ABSTRACT

During acute reperfusion, the expression profiles of long noncoding RNAs in adult rats with focal cerebral ischemia undergo broad changes. However, whether long noncoding RNAs are involved in neuroprotective effects following focal ischemic stroke in rats remains unclear. In this study, RNA isolation and library preparation was performed for long noncoding RNA sequencing, followed by determining the coding potential of identified long noncoding RNAs and target gene prediction. Differential expression analysis, long noncoding RNA functional enrichment analysis, and co-expression network analysis were performed comparing ischemic rats with and without ischemic postconditioning rats. Rats were subjected to ischemic postconditioning via the brief and repeated occlusion of the middle cerebral artery or femoral artery. Quantitative real-time reverse transcription-polymerase chain reaction was used to detect the expression levels of differentially expressed long noncoding RNAs after ischemic postconditioning in a rat model of ischemic stroke. The results showed that ischemic postconditioning greatly affected the expression profile of long noncoding RNAs and mRNAs in the brains of rats that underwent ischemic stroke. The predicted target genes of some of the identified long noncoding RNAs (cis targets) were related to the cellular response to ischemia and stress, cytokine signal transduction, inflammation, and apoptosis signal transduction pathways. In addition, 15 significantly differentially expressed long noncoding RNAs were identified in the brains of rats subjected to ischemic postconditioning. Nine candidate long noncoding RNAs that may be related to ischemic postconditioning were identified by a long noncoding RNA expression profile and long noncoding RNA-mRNA co-expression network analysis. Expression levels were verified by quantitative real-time reverse transcription-polymerase chain reaction. These results suggested that the identified long noncoding RNAs may be involved in the neuroprotective effects associated with ischemic postconditioning following ischemic stroke. The experimental animal procedures were approved by the Animal Experiment Ethics Committee of Kunming Medical University (approval No. KMMU2018018) in January 2018.

6.
Biosci Rep ; 42(1)2022 01 28.
Article in English | MEDLINE | ID: mdl-34897384

ABSTRACT

Treatment of peripheral nerve regeneration with stem cells (SCs) alone has some limitations. For this reason, we evaluate the efficacy of neurotrophic factors combined with stem cell transplantation in the treatment of sciatic nerve injury (SNI) in rats. PubMed, Cochrane Library, Embase, WanFang, VIP and China National Knowledge Infrastructure databases were retrieved from inception to October 2021, and control experiments on neurotrophic factors combined with stem cells in the treatment of SNI in rats were searched. Nine articles and 551 rats were included in the meta-analysis. The results of meta-analysis confirmed that neurotrophic factor combined with stem cells for the treatment of SNI yielded more effective repair than normal rats with regard to sciatic nerve index, electrophysiological detection index, electron microscope observation index, and recovery rate of muscle wet weight. The conclusion is that neurotrophic factor combined with stem cells is more conducive to peripheral nerve regeneration and functional recovery than stem cells alone. However, due to the limitation of the quality of the included literature, the above conclusions need to be verified by randomized controlled experiments with higher quality and larger samples.


Subject(s)
Nerve Regeneration , Sciatic Nerve , Animals , Nerve Growth Factors , Rats , Stem Cell Transplantation
7.
Biomed Pharmacother ; 144: 112273, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34700232

ABSTRACT

Neurogenesis in the adult brain is well recognized and plays a critical role in the maintenance of brain function and homeostasis. However, whether neurogenesis also occurs in the adult peripheral nervous system remains unknown. Here, using sensory ganglia (dorsal root ganglia, DRGs) as a model, we show that neurogenesis also occurs in the peripheral nervous system, but in a manner different from that in the central nervous system. Satellite glial cells (SGCs) express the neuronal precursor markers Nestin, POU domain, class 4, transcription factor 1, and p75 pan-neurotrophin receptor. Following sciatic nerve injury, the suppression of endogenous proBDNF by proBDNF antibodies resulted in the transformation of proliferating SGCs into doublecortin-positive cells in the DRGs. Using purified SGCs migrating out from the DRGs, the inhibition of endogenous proBDNF promoted the conversion of SGCs into neuronal phenotypes in vitro. Our findings suggest that SGCs are neuronal precursors, and that proBDNF maintains the SGC phenotype. Furthermore, the suppression of proBDNF signaling is necessary for neuronal phenotype acquisition by SGCs. Thus, we propose that peripheral neurogenesis may occur via the direct conversion of SGCs into neurons, and that this process is negatively regulated by proBDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Ganglia, Spinal/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Neuroglia/metabolism , Peripheral Nerve Injuries/metabolism , Protein Precursors/metabolism , Action Potentials , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/genetics , Cell Transdifferentiation , Cells, Cultured , Disease Models, Animal , Doublecortin Protein/metabolism , Female , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Male , Neural Stem Cells/pathology , Neuroglia/pathology , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Phenotype , Protein Precursors/genetics , Rats, Sprague-Dawley , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction
8.
Ann Transl Med ; 9(22): 1694, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34988203

ABSTRACT

BACKGROUND: The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is an important mediator of neuroinflammatory responses that regulates inflammatory injury following cerebral ischemia and may be a potential target. Salidroside (Sal) has good anti-inflammatory effects; however, it remains unclear whether Sal can regulate NLRP3 inflammasome activation through the Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway after cerebral ischemia to alleviate inflammatory injury. METHODS: We established an oxygen-glucose deprivation and reoxygenation (OGD/R) model of BV2 cells and a middle cerebral artery occlusion/reperfusion (MCAO/R) rat model. Cell Counting Kit-8 (CCK-8), flow cytometry and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay were used to detect the viability and apoptosis of BV2 cells. Enzyme-linked immunosorbent assay (ELISA) was used to detect the level of inflammatory factors. 2,3,5-triphenyltetrazolium chloride (TTC) staining and modified Neurological Severity Score (mNSS) were used to detect cerebral infarction volume and neurological deficit in rats. Western blot, immunohistochemistry and immunofluorescence staining were used to detect the protein expression levels. RESULTS: Our results showed that Sal increased viability, inhibited lactate dehydrogenase (LDH) release, and reduced apoptosis in OGD/R-induced BV2 cells. Sal reduced the levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-8. Following induction by OGD/R, BV2 cells exhibited NLRP3 inflammasome activation and increased protein levels of NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, IL-1ß, and IL-18. Protein levels of key TLR4 signaling pathway elements, such as TLR4, myeloid differentiation primary response 88 (MyD88), and phosphorylated nuclear factor kappa B p65 (p-NF-κB p65)/NF-κB p65 were upregulated. Interestingly, it was revealed that Sal could reverse these changes. In addition, TAK242, a specific inhibitor of TLR4, had the same effect as Sal treatment on BV2 cells following induction by OGD/R. In the MCAO/R rat model, Sal was also observed to inhibit NLRP3 inflammasome activation in microglia, reduce cerebral infarction volume, and inhibit apoptosis. CONCLUSIONS: In summary, we found that Sal inhibited NLRP3 inflammasome activation and apoptosis in microglia induced by cerebral ischemia/reperfusion injury by inhibiting the TLR4/NF-κB signaling pathway, thus playing a protective role. Therefore, Sal may be a promising drug for the clinical treatment of ischemic stroke.

9.
Metab Brain Dis ; 36(1): 53-65, 2021 01.
Article in English | MEDLINE | ID: mdl-33044640

ABSTRACT

Ischemic postconditioning (PostC) conventionally refers to a series of brief blood vessel occlusions and reperfusions, which can induce an endogenous neuroprotective effect and reduce cerebral ischemia/reperfusion (I/R) injury. Depending on the site of adaptive ischemic intervention, PostC can be classified as in situ ischemic postconditioning (ISPostC) and remote ischemic postconditioning (RIPostC). Many studies have shown that ISPostC and RIPostC can reduce cerebral IS injury through protective mechanisms that increase cerebral blood flow after reperfusion, decrease antioxidant stress and anti-neuronal apoptosis, reduce brain edema, and regulate autophagy as well as Akt, MAPK, PKC, and KATP channel cell signaling pathways. However, few studies have compared the intervention methods, protective mechanisms, and cell signaling pathways of ISPostC and RIPostC interventions. Thus, in this article, we compare the history, common intervention methods, neuroprotective mechanisms, and cell signaling pathways of ISPostC and RIPostC.


Subject(s)
Brain/blood supply , Ischemic Postconditioning/methods , Neuroprotection , Reperfusion Injury/prevention & control , Animals , Disease Models, Animal , Signal Transduction/physiology
10.
J Neurosci Methods ; 346: 108921, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32888963

ABSTRACT

BACKGROUND: Many studies have confirmed that "in situ ischemia postconditioning" (ISPostC) and "remote ischemic postconditioning" (RIPostC) can reduce cerebral ischemia/reperfusion injury, but there is no comparison was made on the consistency of neuroprotection in ISPostC and RIPostC to different ischemic duration and number of cycles. NEW METHOD: We used a transient middle cerebral artery occlusion model to compare the neuroprotection of ISPostC and RIPostC. We conducted ISPostC and RIPostC via brief and repeated MCA and Femoral artery occlusion followed by different ischemic duration and number of cycles. Infarct volume, brain edema, Neurological deficit scores and Apoptosis were evaluated. RESULTS: First, the ISPostC with three cycles of 10-s occlusion/30-s release of both carotid arteries and the RIPostC with three cycles of 10-min occlusion/10-min release of the left and right femoral arteries can obviously reduce cerebral infarction size, brain edema, apoptosis, and improve behavioral deficits than other approaches. Second, three cycles of ischemia/reperfusion may be the best for RIPostC. COMPARISON WITH EXISTING METHOD(S): In this paper, we compared different ischemic duration and frequency of ISPostC and RIPostC models to determine the best method. This conclusion helps to unify the experimental methods. CONCLUSIONS: Different ischemic duration and frequency of ischemic postconditioning affect neuroprotection. three cycles of 10-s occlusion/30-s release of both carotid arteries and three cycles of 10-min occlusion/10-min release of both femoral arteries could be the first choice to study mechanisms of ischemic postconditioning and be conducive to the unification of research results.


Subject(s)
Brain Ischemia , Ischemic Postconditioning , Ischemic Stroke , Stroke , Animals , Infarction, Middle Cerebral Artery/therapy , Neuroprotection , Rats , Rats, Sprague-Dawley
11.
Transl Neurosci ; 11: 17-27, 2020.
Article in English | MEDLINE | ID: mdl-32161682

ABSTRACT

Neuroglial cells have a high level of plasticity, and many types of these cells are present in the nervous system. Neuroglial cells provide diverse therapeutic targets for neurological diseases and injury repair. Cell reprogramming technology provides an efficient pathway for cell transformation during neural regeneration, while transcription factor-mediated reprogramming can facilitate the understanding of how neuroglial cells mature into functional neurons and promote neurological function recovery.

12.
CNS Neurol Disord Drug Targets ; 19(1): 66-82, 2020.
Article in English | MEDLINE | ID: mdl-31957620

ABSTRACT

BACKGROUND: Brain-Derived Neurotrophic Factor (BDNF) plays critical roles during development of the central and peripheral nervous systems, as well as in neuronal survival after injury. Although proBDNF induces neuronal apoptosis after injury in vivo, whether it can also act as a death factor in vitro and in vivo under physiological conditions and after nerve injury, as well as its mechanism of inducing apoptosis, is still unclear. OBJECTIVE: In this study, we investigated the mechanisms by which proBDNF causes apoptosis in sensory neurons and Satellite Glial Cells (SGCs) in Dorsal Root Ganglia (DRG) After Sciatic Nerve Transection (SNT). METHODS: SGCs cultures were prepared and a scratch model was established to analyze the role of proBDNF in sensory neurons and SGCs in DRG following SNT. Following treatment with proBDNF antiserum, TUNEL and immunohistochemistry staining were used to detect the expression of Glial Fibrillary Acidic Protein (GFAP) and Calcitonin Gene-Related Peptide (CGRP) in DRG tissue; immunocytochemistry and Cell Counting Kit-8 (CCK8) assay were used to detect GFAP expression and cell viability of SGCs, respectively. RT-qPCR, western blot, and ELISA were used to measure mRNA and protein levels, respectively, of key factors in BDNF-TrkB, proBDNF-p75NTR/sortilin, and apoptosis signaling pathways. RESULTS: proBDNF induced mitochondrial apoptosis of SGCs and neurons by modulating BDNF-TrkB and proBDNF-p75NTR/sortilin signaling pathways. In addition, neuroprotection was achieved by inhibiting the biological activity of endogenous proBDNF protein by injection of anti-proBDNF serum. Furthermore, the anti-proBDNF serum inhibited the activation of SGCs and promoted their proliferation. CONCLUSION: proBDNF induced apoptosis in SGCs and sensory neurons in DRG following SNT. The proBDNF signaling pathway is a potential novel therapeutic target for reducing sensory neuron and SGCs loss following peripheral nerve injury.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Apoptosis/physiology , Brain-Derived Neurotrophic Factor/metabolism , Ganglia, Spinal/metabolism , Membrane Glycoproteins/metabolism , Receptor, trkB/metabolism , Sciatic Nerve/physiology , Animals , Blotting, Western , Cell Survival , In Situ Nick-End Labeling , Mitochondria/metabolism , Nerve Tissue Proteins , Receptors, Nerve Growth Factor , Sensory Receptor Cells , Signal Transduction
13.
CNS Neurol Disord Drug Targets ; 17(7): 547-556, 2018.
Article in English | MEDLINE | ID: mdl-29992896

ABSTRACT

BACKGROUND & OBJECTIVE: Notoginsenoside R1 (NGR1) is one of the main effective components of Panax notoginseng. METHOD: Primary cortical neurons were harvested from neonatal rats and cultured to analyze the role of NGR1 in neuronal growth and the effects of NGR1 on the Wnt/ß-catenin signaling pathway. Following treatment with NGR1, immunocytochemistry was used to detect expression of Tuj1 and MAP2, and RT-qPCR was used to measure mRNA levels of key factors in the Wnt signaling pathway. RESULTS: Results showed that NGR1 promotes growth of cultured neurons and significantly upregulates mRNA levels of ß-catenin, Dishevelled, and Frizzled. To further confirm whether NGR1 promoted cortical neuron growth via the Wnt/ß-catenin signaling pathway, we knocked down ß- catenin mRNA by siRNA interference; following NGR1 treatment of ß-catenin-knockdown neurons, ß-catenin mRNA levels increased significantly. CONCLUSION: In conclusion, these results demonstrate that NGR1 promotes growth of cultured cortical neurons from the neonatal rat, possibly via the Wnt/ß-catenin signaling pathway.


Subject(s)
Cerebral Cortex/cytology , Ginsenosides/pharmacology , Neurons/drug effects , beta Catenin/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Ginsenosides/genetics , Ginsenosides/metabolism , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Patch-Clamp Techniques , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Transfection , Tubulin/genetics , Tubulin/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...