Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Affiliation country
Publication year range
1.
J Neuroinflammation ; 21(1): 85, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38582897

ABSTRACT

Homer1a and A2 astrocytes are involved in the regulation of inflammation induced by intracerebral hemorrhage (ICH). However, there is no anticipated treatment strategy based on the anti-inflammatory effect of Homer1a and A2 astrocytes. Here, we successfully induced A2 astrocytes in vitro, and then we report an efficient method to prepare Homer1a+ EVs derived from A2 astrocytes which making it more stable, safe, and targetable to injured neurons. Homer1a+ EVs promotes the conversion of A1 to A2 astrocytes in ICH mice. Homer1a+ EVs inhibits activation and nuclear translocation of NF-κB, thereby regulating transcription of IL-17A in neurons. Homer1a+ EVs inhibits the RAGE/NF-κB/IL-17 signaling pathway and the binding ability of IL-17A: IL17-AR and RAGE: DIAPH1. In addition, Homer1a+ EVs ameliorates the pathology, behavior, and survival rate in GFAPCreHomer1fl/-Homer1a± and NestinCreRAGEfl/fl ICH mice. Our study provides a novel insight and potential for the clinical translation of Homer1a+ EVs in the treatment of ICH.


Subject(s)
Extracellular Vesicles , NF-kappa B , Mice , Animals , NF-kappa B/metabolism , Interleukin-17 , Cerebral Hemorrhage/metabolism , Signal Transduction , Extracellular Vesicles/metabolism
2.
Inflamm Res ; 73(1): 131-144, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38091015

ABSTRACT

OBJECTIVE: Proinflammatory necroptosis is the main pathological mechanism of ischemic stroke. Homer scaffolding protein 1 (Homer1) is a postsynaptic scaffolding protein that exerts anti-inflammatory effects in most central nervous system diseases. However, the relationship between Homer1 and proinflammatory necroptosis in ischemic stroke remains unclear. AIM: This study aimed to investigate the role of Homer1 in ischemia-induced necroptosis. METHODS: C57BL/6 mice were used to establish a model of permanent middle cerebral artery occlusion model (pMCAO). Homer1 knockdown mice were generated using adeno-associated virus (AAV) infection to explore the role of Homer1 and its impact on necroptosis in pMCAO. Finally, Homer1 protein was stereotaxically injected into the ischemic cortex of Homer1flox/flox/Nestin-Cre +/- mice, and the efficacy of Homer1 was investigated using behavioral assays and molecular biological assays to explore potential mechanisms. RESULTS: Homer1 expression peaked at 8 h in the ischemic penumbral cortex after pMCAO and colocalized with neurons. Homer1 knockdown promoted neuronal death by enhancing necroptotic signaling pathways and aggravating ischemic brain damage in mice. Furthermore, the knockdown of Homer1 enhanced the expression of proinflammatory cytokines. Moreover, injection of Homer1 protein reduced necroptosis-induced brain injury inhibited the expression of proinflammatory factors, and ameliorated the outcomes in the Homer1flox/flox/Nestin-Cre+/- mice after pMCAO. CONCLUSIONS: Homer1 ameliorates ischemic stroke by inhibiting necroptosis-induced neuronal damage and neuroinflammation. These data suggested that Homer1 is a novel regulator of neuronal death and neuroinflammation.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Mice , Animals , Ischemic Stroke/complications , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Nestin/metabolism , Nestin/pharmacology , Neuroinflammatory Diseases , Necroptosis , Mice, Inbred C57BL , Infarction, Middle Cerebral Artery/pathology , Neurons/pathology , Brain Ischemia/metabolism , Stroke/complications , Stroke/metabolism , Stroke/pathology , Homer Scaffolding Proteins/genetics , Homer Scaffolding Proteins/metabolism , Homer Scaffolding Proteins/pharmacology
3.
J Neuroinflammation ; 19(1): 67, 2022 Mar 14.
Article in English | MEDLINE | ID: mdl-35287697

ABSTRACT

BACKGROUND: Inflammation induced by intracerebral hemorrhage (ICH) is one of the main causes of the high mortality and poor prognosis of patients with ICH. A1 astrocytes are closely associated with neuroinflammation and neurotoxicity, whereas A2 astrocytes are neuroprotective. Homer scaffolding protein 1 (Homer1) plays a protective role in ischemic encephalopathy and neurodegenerative diseases. However, the role of Homer1 in ICH-induced inflammation and the effect of Homer1 on the phenotypic conversion of astrocytes remain unknown. METHODS: Femoral artery autologous blood from C57BL/6 mice was used to create an ICH model. We use the A1 phenotype marker C3 and A2 phenotype marker S100A10 to detect astrocyte conversion after ICH. Homer1 overexpression/knock-down mice were constructed by adeno-associated virus (AAV) infection to explore the role of Homer1 and its mechanism of action after ICH. Finally, Homer1 protein and selumetinib were injected into in situ hemorrhage sites in the brains of Homer1flox/flox/Nestin-Cre+/- mice to study the efficacy of Homer1 in the treatment of ICH by using a mouse cytokine array to explore the potential mechanism. RESULTS: The expression of Homer1 peaked on the third day after ICH and colocalized with astrocytes. Homer1 promotes A1 phenotypic conversion in astrocytes in vivo and in vitro. Overexpression of Homer1 inhibits the activation of MAPK signaling, whereas Homer1 knock-down increases the expression of pathway-related proteins. The Homer1 protein and selumetinib, a non-ATP competitive MEK1/2 inhibitor, improved the outcome in ICH in Homer1flox/flox/Nestin-Cre+/- mice. The efficacy of Homer1 in the treatment of ICH is associated with reduced expression of the inflammatory factor TNFSF10 and increased expression of the anti-inflammatory factors activin A, persephin, and TWEAK. CONCLUSIONS: Homer1 plays an important role in inhibiting inflammation after ICH by suppressing the A1 phenotype conversion in astrocytes. In situ injection of Homer1 protein may be a novel and effective method for the treatment of inflammation after ICH.


Subject(s)
Astrocytes , Cerebral Hemorrhage , Animals , Astrocytes/metabolism , Cerebral Hemorrhage/metabolism , Homer Scaffolding Proteins/genetics , Homer Scaffolding Proteins/metabolism , Homer Scaffolding Proteins/pharmacology , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Biochem Biophys Res Commun ; 512(4): 763-769, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30926163

ABSTRACT

EP300-interacting inhibitor of differentiation 1 (Eid1) regulates differentiation, transcription and acetyltransferase activity. But the main function of Eid1 in the brain is still unclear. To better understand this issue, we generated Eid1-knockout (Eid1-KO) mice. We found poorer learning and memory ability, and smaller volume of neonatal telencephalon in Eid1-KO group than wild-type (WT). Bioinformatics implied that Eid1 may directly regulate cell proliferation. We then isolated neural stem cells (NSCs) and discovered a slower proliferation rate in Eid1-KO NSCs. Moreover, based on bioinformatics results, we investigated the expression of phosphatidylinositol 3-kinase (PI3K)/AKT/GSK3ß pathway by Western blotting assay, which showed attenuated in Eid1-KO group. Our data proved the first comprehensive report of Eid1 regulating NSCs proliferation via PI3K/AKT/GSK3ß pathway, and provide a foundation for the role of EID1 in the brain.


Subject(s)
Cell Proliferation , Neural Stem Cells/cytology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/metabolism , Nuclear Proteins/genetics , Repressor Proteins/genetics , Signal Transduction
5.
J Neuroinflammation ; 16(1): 234, 2019 Nov 26.
Article in English | MEDLINE | ID: mdl-31771613

ABSTRACT

BACKGROUND: Inflammation and apoptosis caused by intracerebral hemorrhage (ICH) are two important factors that affect patient prognosis and survival. Toll-like receptor 4 (TLR4) triggers activation of the inflammatory pathway, causing synthesis and release of inflammatory factors. The inflammatory environment also causes neuronal apoptosis. However, no studies have reported the role of TLR4 in inflammation and apoptosis. METHODS: We performed survival curve analysis and behavioral scores on TLR4 knockout mice and wild-type mice after inducing ICH. We used TLR4 knockout mice and wild-type mice to make ICH models with type VII collagenase and explored the link between TLR4 in inflammation and apoptosis. We used Western blot to detect the expression of apoptosis-related proteins, inflammatory factors, and their receptors at different time points after ICH induction. The effects of TLR4 on apoptosis were observed by TUNEL, Hoechst, and HE staining techniques. The association with TLR4 in inflammation and apoptosis was explored using IL-1ß and TNF-α antagonists. Data conforming to a normal distribution are expressed as mean ± standard deviation. Grade and quantitative data were compared with rank sum test and t test between two groups. P < 0.05 was considered statistically significant. RESULTS: TLR4 knockout significantly increased the survival rate of ICH mice. The scores of TLR4 knockout mice were significantly lower than those of wild-type mice. We found that TLR4 knockout mice significantly inhibited apoptosis and the expression of inflammatory factors after the induction of ICH. The apoptosis of ICH-induced mice was significantly improved after injecting IL-1ß and TNF-α antagonists. Moreover, the anti-apoptotic effect of the antagonist in wild-type mice is more pronounced. A single injection of the antagonist failed to improve apoptosis in TLR4 knockout mice. CONCLUSIONS: We conclude that TLR4-induced inflammation after ICH promotes neuronal apoptosis. IL-1ß and TNF-α antagonists attenuate this apoptotic effect. Therefore, targeting TLR4 in patients with clinical ICH may attenuate inflammatory response, thereby attenuating apoptosis and improving prognosis.


Subject(s)
Apoptosis/physiology , Brain/metabolism , Cerebral Hemorrhage/metabolism , Toll-Like Receptor 4/metabolism , Animals , Apoptosis/drug effects , Brain/drug effects , Cerebral Hemorrhage/genetics , Collagenases/metabolism , Interleukin-1beta/antagonists & inhibitors , Mice , Mice, Knockout , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors
6.
J Inflamm Res ; 17: 1337-1347, 2024.
Article in English | MEDLINE | ID: mdl-38434583

ABSTRACT

Purpose: We aim to explore the relationship between Homer1 and the outcomes of AIS patients at 3 months. Patients and Methods: This prospective cohort study was conducted from May 2022 to March 2023. In this study, we investigated the association between serum Homer1 levels by enzyme-linked immunosorbent assay at admission and functional outcomes of patients at 3 months after AIS. Results: Overall, 89 AIS patients (48 good outcomes and 41 poor outcomes) and 83 healthy controls were included. The median serum Homer1 level of patients at admission with poor outcomes was significantly higher than that of patients with good outcomes (39.33 vs 33.15, P<0.001). Serum Homer1 levels at admission were positively correlated with the severity of AIS (r = 0.488, P<0.001). The optimal cutoff of serum Homer1 level as an indicator for an auxiliary diagnosis of 3 months functional outcomes was 35.07 pg/mL, with a sensitivity of 75.0% and a specificity of 92.7% (AUC 0.837; 95% CI [0.744-0.907]; P<0 0.001). The odds ratio of MRS > 2 predicted by the level of serum Homer1 after 3 months was 1.665 (1.306-2.122; P<0.001). Conclusion: Serum concentrations of Homer1 have a high predictive value for neurobehavioral outcomes after acute ischemic stroke. Higher serum Homer1 levels (>35.07 pg/mL) were positively associated with poor functional outcomes of patients 3 months post-stroke.

7.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167435, 2024 10.
Article in English | MEDLINE | ID: mdl-39067535

ABSTRACT

INTRODUCTION: Microglia play pivotal roles in post-intracerebral hemorrhage (ICH) neural injury. Iron metabolism, which is dysregulated after ICH, participates in microglial dysfunction. Previous studies have shown that iron metabolism-related lipocalin-2 (LCN2) is involved in regulating microglial function following ICH. In this study, we investigated the role of LCN2 in microglial function following ICH. METHODS: The BV2 (microglia) cell line, transfected with LCN2 for overexpression/interference, received a blood infusion from C57BL/6 mice in vitro. For the in vivo study of LCN2 function, an LCN2 knockout was conducted in mice. Liproxstatin-1 and RSL3 were used to manipulate ferroptosis and to study the effects of LCN2 on microglia after ICH. A BV2 (microglia) cell line, transfected with ferritin light chain (FTL) for overexpression/interference, was co-cultured with primary cultured neurons for a study on the mechanism of LCN2. Behavioral tests were conducted pre-ICH and on days 3, 7, and 28 post-ICH, and the brains and cultured cells were collected for protein, histological, and morphological studies. RESULTS: Brain LCN2 expression was upregulated in microglia, astrocytes, and neurons and played hazardous roles after ICH. In microglia, LCN2 promoted ferroptosis, which facilitated neural injury after ICH. LCN2-mediated FTL deficiency was shown to be responsible for microglial ferroptosis-induced neural injury. CONCLUSION: Our study suggests that LCN2-enhanced microglial ferroptosis plays a detrimental role by inducing FTL deficiency after ICH. The current study reveals a novel molecular mechanism involved in the pathophysiological progression of ICH.


Subject(s)
Cerebral Hemorrhage , Ferroptosis , Lipocalin-2 , Mice, Knockout , Microglia , Animals , Lipocalin-2/metabolism , Lipocalin-2/genetics , Microglia/metabolism , Microglia/pathology , Microglia/drug effects , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/genetics , Ferroptosis/drug effects , Mice , Male , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Neurons/drug effects , Apoferritins/metabolism , Apoferritins/genetics , Disease Models, Animal , Cell Line
8.
Food Chem ; 426: 136645, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37379695

ABSTRACT

In this work, Ti3C2 nano-enzymes (Ti3C2 NEs) materials with simulated peroxidase activity and fluorescence quenching properties were prepared. Then Ti3C2 NEs was functionalized using 6-carboxyfluorescein (FAM) labeled Aflatoxin B1 (AFB1) aptamers to construct a novel multimode nano enzyme biosensor for the detection of AFB1 in peanuts. Based on the fluorescence quenching characteristics and the superior simulated peroxidase activity of Ti3C2 NES and the specific binding of the aptamer to AFB1, the sensitive and rapid fluorescence/colorimetric/smart phone detection of AFB1 have been achieved, with detection limits of 0.09 ng mL-1, 0.61 ng mL-1 and 0.96 ng mL-1, respectively. The analytical method provided can not only detect AFB1 in multiple modes, but also has a wider detection range, lower limit of detection (LOD) and better recovery rate, and can achieve on-site accurate detection of AFB1 content in peanuts, which has great application potential in the field of food quality testing.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Aptamers, Nucleotide/chemistry , Food , Aflatoxin B1/analysis , Arachis , Biosensing Techniques/methods , Peroxidases , Limit of Detection , Food Contamination/analysis
9.
Exp Mol Med ; 55(6): 1203-1217, 2023 06.
Article in English | MEDLINE | ID: mdl-37258577

ABSTRACT

The tripartite motif (TRIM) 22 and mitogen-activated protein kinase (MAPK) signaling pathways play critical roles in the growth of glioblastoma (GBM). However, the molecular mechanism underlying the relationship between TRIM22 and MAPK signaling remains unclear. Here, we found that TRIM22 binds to exon 2 of the sphingosine kinase 2 (SPHK2) gene. An ERK1/2-driven luciferase reporter construct identified TRIM22 as a potential activator of MAPK signaling. Knockout and overexpression of TRIM22 regulate the inhibition and activation of MAPK signaling through the RING-finger domain. TRIM22 binds to Raf-1, a negative regulator of MAPK signaling, and accelerates its degradation by inducing K48-linked ubiquitination, which is related to the CC and SPRY domains of TRIM22 and the C1D domain of Raf-1. In vitro and in vivo, an SPHK2 inhibitor (K145), an ERK1/2 inhibitor (selumetinib), and the nonphosphorylated mutant Raf-1S338A inhibited GBM growth. In addition, deletion of the RING domain and the nuclear localization sequence of TRIM22 significantly inhibited TRIM22-induced proliferation of GBM cells in vivo and in vitro. In conclusion, our study showed that TRIM22 regulates SPHK2 transcription and activates MAPK signaling through posttranslational modification of two critical regulators of MAPK signaling in GBM cells.


Subject(s)
Glioblastoma , Mitogen-Activated Protein Kinases , Humans , Mitogen-Activated Protein Kinases/metabolism , Glioblastoma/genetics , Signal Transduction , Cell Line , Cell Proliferation , Minor Histocompatibility Antigens , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Repressor Proteins/genetics
10.
Neural Regen Res ; 18(8): 1734-1742, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36751799

ABSTRACT

Intracerebral hemorrhage is often accompanied by oxidative stress induced by reactive oxygen species, which causes abnormal mitochondrial function and secondary reactive oxygen species generation. This creates a vicious cycle leading to reactive oxygen species accumulation, resulting in progression of the pathological process. Therefore, breaking the cycle to inhibit reactive oxygen species accumulation is critical for reducing neuronal death after intracerebral hemorrhage. Our previous study found that increased expression of nicotinamide adenine dinucleotide phosphate oxidase 4 (NADPH oxidase 4, NOX4) led to neuronal apoptosis and damage to the blood-brain barrier after intracerebral hemorrhage. The purpose of this study was to investigate the role of NOX4 in the circle involving the neuronal tolerance to oxidative stress, mitochondrial reactive oxygen species and modes of neuronal death other than apoptosis after intracerebral hemorrhage. We found that NOX4 knockdown by adeno-associated virus (AAV-NOX4) in rats enhanced neuronal tolerance to oxidative stress, enabling them to better resist the oxidative stress caused by intracerebral hemorrhage. Knockdown of NOX4 also reduced the production of reactive oxygen species in the mitochondria, relieved mitochondrial damage, prevented secondary reactive oxygen species accumulation, reduced neuronal pyroptosis and contributed to relieving secondary brain injury after intracerebral hemorrhage in rats. Finally, we used a mitochondria-targeted superoxide dismutase mimetic to explore the relationship between reactive oxygen species and NOX4. The mitochondria-targeted superoxide dismutase mimetic inhibited the expression of NOX4 and neuronal pyroptosis, which is similar to the effect of AAV-NOX4. This indicates that NOX4 is likely to be an important target for inhibiting mitochondrial reactive oxygen species production, and NOX4 inhibitors can be used to alleviate oxidative stress response induced by intracerebral hemorrhage.

11.
Biofabrication ; 15(2)2023 03 14.
Article in English | MEDLINE | ID: mdl-36812580

ABSTRACT

Although autologous bone (AB) grafting is considered to be the gold standard for cranioplasty, unresolved problems remain, such as surgical-site infections and bone flap absorption. In this study, an AB scaffold was constructed via three-dimensional (3D) bedside-bioprinting technology and used for cranioplasty. To simulate the skull structure, a polycaprolactone shell was designed as an external lamina, and 3D-printed AB and a bone marrow-derived mesenchymal stem cell (BMSC) hydrogel was used to mimic cancellous bone for bone regeneration. Ourin vitroresults showed that the scaffold exhibited excellent cellular affinity and promoted osteogenic differentiation of BMSCs in both two-dimensional and 3D culture systems. The scaffold was implanted in beagle dog cranial defects for up to 9 months, and the scaffold promoted new bone and osteoid formation. Furtherin vivostudies indicated that transplanted BMSCs differentiated into vascular endothelium, cartilage, and bone tissues, whereas native BMSCs were recruited into the defect. The results of this study provide a method for bedside bioprinting of a cranioplasty scaffold for bone regeneration, which opens up another window for clinical applications of 3D printing in the future.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Animals , Dogs , Tissue Scaffolds/chemistry , Bone Regeneration , Cell Differentiation , Skull/surgery , Printing, Three-Dimensional , Tissue Engineering/methods
12.
Neuroscience ; 480: 97-107, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34798181

ABSTRACT

Ischemic injury in patients with stroke often leads to neuronal damage and mitochondrial dysfunction. Neuronal injury caused by ischemia can be partly attributed to glutamate (L-Glu) excitotoxicity. Previous studies have shown that PTEN-induced kinase 1 (PINK1) plays a neuroprotective role in ischemic brain injury by regulating mitochondrial integrity and function. However, there are few reports on the expression of PINK1 in L-Glu excitotoxicity models, its effect on neuronal survival, and whether PINK1 plays a protective role in stroke by regulating mitophagy. In the present study, different concentrations of L-Glu inhibited the viability of neurons. After L-Glu treatment at different times, the mRNA level, protein level, and cellular fluorescence intensity of PINK1 first increased and then decreased. Compared with normal cells, cells with low PINK1 expression enhanced the inhibitory effect of L-Glu on neuronal activity, while those with high PINK1 expression showed a protective effect on neurons by alleviating mitochondrial membrane potential loss. In addition, RAP (an autophagy activator) could increase the co-localization of the mitophagy-related proteins light chain 3 (LC3) and Tom20, whereas 3-MA (an autophagy inhibitor) exerted the opposite effect. Finally, we found that L-Glu could induce the expression of PINK1/Parkin/ LC3 in neurons at both mRNA and protein levels, while RAP could further increase their expression, and 3-MA decreased their expression. Taken together, PINK1 protects against L-Glu-induced neuronal injury by protecting mitochondrial function, and the potential protective mechanism may be closely related to the enhancement of mitophagy mediated by the PINK1/Parkin signaling pathway.


Subject(s)
Neuroprotective Agents , Glutamic Acid/toxicity , Humans , Mitophagy , Neurons , Neuroprotective Agents/pharmacology , Protein Kinases/pharmacology , Ubiquitin-Protein Ligases
13.
J Oncol ; 2022: 9499317, 2022.
Article in English | MEDLINE | ID: mdl-39280892

ABSTRACT

Low-grade gliomas (LGGs) are primary invasive brain tumors that grow slowly but are incurable and eventually develop into high malignant glioma. Fc fragment of IgG receptor IIIa (FCGR3A) gene polymorphism may correlate with some cancers' treatment responses. However, the expression and prognosis value of FCGR3A and correlation with tumor-immune infiltrate in LGG remain unclear. FCGR3A mRNA expression in gastric cancer (GC) was examined using TIMER and GEPIA databases. Correlations between FCGR3A expression and clinicopathological parameters were analyzed using ULACAN and CGGA databases. GEPIA, OncoLnc, and ULACAN databases were used to examine the clinical prognostic significance of FCGR3A in LGG. TIMER was used to analyze the correlations among FCGR3A and tumor-infiltrating immune cells. Signaling pathways related to FCGR3A expression were identified by LinkedOmics. We found that FCGR3A expression was higher in LGG than in normal tissue and was correlated with various clinical parameters. In addition, high FCGR3A expression predicted poor overall survival in LGG. More importantly, FCGR3A expression positively correlated with immune checkpoint molecules, including PD1, PD-L1, PD-L2, CTLA4, LAG-3 and TIM-3, and tumor-associated macrophage (TAM) gene markers in LGG. GO and KEGG pathway analyses indicated that TUBA1C may potentially regulate the pathogenesis of LGG through immune-related pathways. These findings indicated that FCGR3A plays a vital role in the infiltration of immune cells and could constitute a promising prognostic biomarker in LGG patients.

14.
Comput Math Methods Med ; 2022: 3436631, 2022.
Article in English | MEDLINE | ID: mdl-35912147

ABSTRACT

Objective: To develop and authenticate a risk stratification framework and nomogram for ascertaining cancer-specific survival (CSS) among the pediatric brainstem gliomas. Methods: For patients less than 12 years, according to Surveillance, Epidemiology, and End Results (SEER), information from 1998 to 2016 is found in their databases. The survival outcomes, treatments, and demographic clinicopathologic conditions are scrutinized per the database validation, and training cohorts are divided and validated using multivariate Cox regression analysis. A nomogram was designed, and predominantly, the risk stratification conceptualization engaged selected tenets according to the multivariate analysis. The model's authenticity was substantiated through C-index measure and calibration curves. Results: There are 806 pediatric concerns of histologically concluded brainstem glioma in the research. According to multivariate analysis, age, grade, radiotherapy, and race (with P value < 0.05) depicted independent prognostic variations of the pediatric gliomas. The nomogram's C-index was approximately 0.75 and an accompanied predictive capability for CSS. Conclusion: The nomogram constructed in this glioma's context is the primary predictor of using risk stratification. A combination of nomograms with the risk stratification mechanism assists clinicians in monitoring high-risk individuals and engage targeted accessory treatment.


Subject(s)
Brain Neoplasms/mortality , Brain Stem/pathology , Glioma/mortality , Brain Neoplasms/therapy , Child , Child, Preschool , Cohort Studies , Glioma/therapy , Humans , Infant , Multivariate Analysis , Nomograms , Prognosis , Regression Analysis , Risk Assessment/methods , SEER Program
15.
Neuroscience ; 492: 1-17, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35405301

ABSTRACT

Toll-like receptor-4 (TLR4), a member of the TLR family, plays a key role in inflammation-related diseases of the nervous system. TLR4 knockout mice are widely used in various neurological disease studies, and there is a clear correlation between inflammation and behavior. Therefore, elucidating the effect of TLR4 on neurobehavioral function is essential, and the related mechanisms need to be explored. Male TLR4 knockout (TLR4-/-) and wild-type (TLR4+/+) mice of different ages (4, 8, and 16 months) were used for behavioral experiments. Synaptic spine, blood-brain barrier (BBB) integrity, memory regulatory proteins, cortical blood flow, and inflammatory factor examinations were also conducted to explore the possible mechanism by which TLR4 works. Here, we found that compared with 16-m-old TLR4+/+ mice, age-matched TLR4-/- mice had better learning and memory abilities, increased expression of neuronal synaptic spines, and increased memory-related regulatory proteins in the hippocampus. TLR4 knockout significantly attenuated the fear response in 16-m-old mice. The TLR4-/- mice also had better blood-brain barrier integrity, increased expression of tight junction-associated proteins, increased cerebral cortical blood flow and reduced proinflammatory cytokine expression in the cortex and cerebrospinal fluid. Our results suggest that TLR4 deletion ameliorates significant neurobehavioral dysfunction during the aging stage, as well as multiple abnormalities in brain function and structure due to alterations in tight junction-associated proteins and inflammatory factors.


Subject(s)
Brain , Toll-Like Receptor 4 , Animals , Brain/metabolism , Cognition , Gene Deletion , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tight Junction Proteins/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
16.
Mol Ther Oncolytics ; 26: 413-428, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36159777

ABSTRACT

Tripartite motif 22 (TRIM22) is an agonist of nuclear factor κB (NF-κB) that plays an important role in the proliferation and drug sensitivity of glioblastoma (GBM). However, the molecular mechanism underlying the protein network between TRIM22 and nuclear factor κB (NF-κB) in GBM remains unclear. Here, we found that knockout of TRIM22 effectively inhibited tumor proliferation and increased the sensitivity of GBM cells to temozolomide (TMZ) in vivo and in vitro. Moreover, TRIM22 forms a complex with cytosolic purine 5-nucleotidase (NT5C2) in GBM and regulates the ubiquitination of retinoic acid-inducible gene-I (RIG-I). TRIM22 promotes the K63-linked ubiquitination of RIG-I, while NT5C2 is responsible for K48-linked ubiquitination. This regulation directly affects the RIG-I/NF-κB/cell division cycle and apoptosis regulator protein 1 (CCAR1) signaling axis. Ubiquitin modification inhibitor of RIG-I restores the inhibition of tumor growth induced by TRIM22 knockout. The follow-up results showed that compared with patients with high TRIM22 expression, patients with low TRIM22 expression had a longer survival time and were more sensitive to treatment with TMZ. Our results revealed that the TRIM22-NT5C2 complex orchestrates the proliferation of GBM and benefits of TMZ through post-translational modification of RIG-I and the regulation of the RIG-I/NF-κB/CCAR1 pathway and is a promising target for single-pathway multi-target therapy.

17.
Chin Neurosurg J ; 8(1): 45, 2022 Dec 30.
Article in English | MEDLINE | ID: mdl-36582003

ABSTRACT

BACKGROUND: Awake craniotomy (AC) has become gold standard in surgical resection of gliomas located in eloquent areas. The conscious sedation techniques in AC include both monitored anesthesia care (MAC) and asleep-awake-asleep (AAA). The choice of optimal anesthetic method depends on the preferences of the surgical team (mainly anesthesiologist and neurosurgeon). The aim of this study was to compare the difference in physiological and blood gas data, dosage of different drugs, the probability of switching to endotracheal intubation, and extent of tumor resection and dysfunction after operation between AAA and MAC anesthetic management for resection of gliomas in eloquent brain areas. METHODS: Two-hundred and twenty-five patients with super-tentorial tumor located in eloquent areas underwent AC from 2009 to 2021 in Xijing Hospital. Forty-one patients underwent AAA technique, and the rest one-hundred eighty-four patients underwent MAC technique. Anesthetic management, dosage of different drugs, intraoperative complications, postoperative outcomes, adverse events, extent of resection and motor, and sensory and language dysfunction after operation were compared between MAC and AAA. RESULT: There was no significant difference in gender, KPS score, MMSE score, glioma grade, type, and growth site between the patients in the two groups, except the older age of patients in MAC group than that in AAA group. During the whole process of operation, there were greater pulse pressure difference (P = 0.046), shorter operation time (P = 0.039), less dosage of remifentanil (P = 0.000), more dosage of dexmedetomidine (P = 0.013), more use of antiemetics (81%, P = 0.0067), lower use of vasoactive agent (45.1%, P = 0.010), and lower probability of conversion to general anesthesia (GA, P = 0.027) in MAC group than that in AAA group. Blood gas analysis showed that PetCO2 (P = 0.000), Glu concentration (P = 0.000), and PaCO2 (P = 0.000) were higher, but SPO2 (P = 0.002) and PaO2 (P = 0.000) were lower in MAC group than that in AAA group. In the postoperative recovery stage, compared with that of AAA group, the probability of dysfunction in MAC group at 1, 3, 5, and 7 days after operation was lower, which were 27.8% vs 53.6% (P = 0.003), 31% vs 68.3% (P = 0.000), 28.8% vs 63.4% (P = 0.000), and 25.6% vs 58.5% (P = 0.000), respectively. CONCLUSION: Compared with AAA, it seems that MAC has more advantages in the management for resection of gliomas in eloquent brain areas, and MAC combined with multiple monitoring such as cerebral cortical mapping, neuronavigation, and ultrasonic detection is worthy of popularization for the resection of gliomas in eloquent brain areas.

18.
Front Oncol ; 12: 803652, 2022.
Article in English | MEDLINE | ID: mdl-36106122

ABSTRACT

Glioblastoma (GBM) is a highly invasive neurological malignancy with poor prognosis. LncRNA-GAS5 (growth arrest-specific transcript 5) is a tumor suppressor involved in multiple cancers. In this study, we explored the clinical significance, biological function, and underlying mechanisms of GAS5 in GBM. We showed that lncRNA-GAS5 expression decreased in high-grade glioma tissues and cells, which might be associated with poor prognosis. GAS5 overexpression lowered cell viability, suppressed GBM cell migration and invasion, and impaired the stemness and proliferation of glioma stem cells (GSCs). We further discovered that GAS5 inhibited the viability of glioma cells through miR-let-7e and miR-125a by protecting SPACA6 from degradation. Moreover, GAS5 played an anti-oncogenic role in GBM through the combined involvement of let-7e and miR-125a in vivo and in vitro. Notably, these two miRNAs block the IL-6/STAT3 pathway in tumor tissues extracted from a xenograft model. Taken together, our study provides evidence for an important role of GAS5 in GBM by affecting the proliferation and migration of GSCs, thus providing a new potential prognostic biomarker and treatment strategy for GBM.

19.
J Cell Physiol ; 226(2): 440-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20672326

ABSTRACT

Brain natriuretic peptide (BNP) may act as a neuromodulator via its associated receptors (natriuretic peptide receptors, NPRs) in the central nervous system (CNS), but few studies have reported its activity in the peripheral nervous system (PNS). In this study, we observed that BNP increased the tetraethylammonium chloride (TEA)-sensitive delayed rectifier outward potassium current (I(K)) in mouse Schwann cells (SCs) using whole-cell recording techniques. At concentrations of 1-100 nM, BNP reversibly activated I(K) in a dose-dependent manner, with modulating its steady-state activation and inactivation properties. The effect of BNP on I(K) was abolished by preincubation with the specific antagonist of NPR-A, and could not be mimicked by application of NPR-C agonist. These results were supported by immunocytochemical findings indicating that NPR-A was expressed in SCs. The application of 8-Br-guanosine 3',5'-monophosphate (8-Br-cGMP) mimicked the effect of BNP on I(K), but BNP was unable to further increase I(K) after the application of cyclic guanosine monophosphate (cGMP). Genistein blocked I(K) and also completely eliminated the effects of BNP and cGMP on I(K). The selective K(V)2.1 subunit blocker, Jingzhaotoxin-III (JZTX-III), reduced I(K) amplitude by 30%, but did not abolish the increase effect of BNP on I(K) amplitude. In addition, BNP significantly stimulated SCs proliferation and this effect could be partly inhibited by TEA. Together these results suggest that BNP modulated I(K) probably via cGMP- and tyrosine kinase-dependent pathways by activation of NPR-A. This effect of BNP on I(K) in SCs might partly explain its effect on cell proliferation.


Subject(s)
Cell Proliferation/drug effects , Delayed Rectifier Potassium Channels/metabolism , Natriuretic Peptide, Brain/pharmacology , Schwann Cells/drug effects , Schwann Cells/physiology , Animals , Cyclic AMP/chemistry , Cyclic AMP/metabolism , Genistein/metabolism , HEK293 Cells , Humans , Membrane Potentials/drug effects , Mice , Patch-Clamp Techniques , Peptides/metabolism , Protein Kinase Inhibitors/metabolism , Protein Subunits/metabolism , Schwann Cells/cytology , Spider Venoms/metabolism , Tetraethylammonium/metabolism
20.
Front Cell Dev Biol ; 9: 679866, 2021.
Article in English | MEDLINE | ID: mdl-34858969

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a common pulmonary complication observed in preterm infants that is composed of multifactorial pathogenesis. Current strategies, albeit successful in moderately reducing morbidity and mortality of BPD, failed to draw overall satisfactory conclusion. Here, using a typical mouse model mimicking hallmarks of BPD, we revealed that both cord blood-derived mononuclear cells (CB-MNCs) and umbilical cord-derived mesenchymal stem cells (UC-MSCs) are efficient in alleviating BPD. Notably, infusion of CB-MNCs has more prominent effects in preventing alveolar simplification and pulmonary vessel loss, restoring pulmonary respiratory functions and balancing inflammatory responses. To further elucidate the underlying mechanisms within the divergent therapeutic effects of UC-MSC and CB-MNC, we systematically investigated the long noncoding RNA (lncRNA)-microRNA (miRNA)-messenger RNA (mRNA) and circular RNA (circRNA)-miRNA-mRNA networks by whole-transcriptome sequencing. Importantly, pathway analysis integrating Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG)/gene set enrichment analysis (GSEA) method indicates that the competing endogenous RNA (ceRNA) network is mainly related to the regulation of GTPase activity (GO: 0043087), extracellular signal-regulated kinase 1 (ERK1) and ERK2 signal cascade (GO: 0070371), chromosome regulation (GO: 0007059), and cell cycle control (GO: 0044770). Through rigorous selection of the lncRNA/circRNA-based ceRNA network, we demonstrated that the hub genes reside in UC-MSC- and CB-MNC-infused networks directed to the function of cell adhesion, motor transportation (Cdk13, Lrrn2), immune homeostasis balance, and autophagy (Homer3, Prkcd) relatively. Our studies illustrate the first comprehensive mRNA-miRNA-lncRNA and mRNA-miRNA-circRNA networks in stem cell-infused BPD model, which will be valuable in identifying reliable biomarkers or therapeutic targets for BPD pathogenesis and shed new light in the priming and conditioning of UC-MSCs or CB-MNCs in the treatment of neonatal lung injury.

SELECTION OF CITATIONS
SEARCH DETAIL