41 results on '"Zhongyuan Bao"'
Search Results
2. Supplemental Figure S6 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S6
- Published
- 2023
- Full Text
- View/download PDF
3. Supplemental Figure S9 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S9
- Published
- 2023
- Full Text
- View/download PDF
4. Supplemental Figure S1 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S1
- Published
- 2023
- Full Text
- View/download PDF
5. Supplemental Figure S3 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S3
- Published
- 2023
- Full Text
- View/download PDF
6. Supplemental Figure S7 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S7
- Published
- 2023
- Full Text
- View/download PDF
7. Supplemental Figure S4 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S4
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure S5 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S5
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental Figure S8 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S8
- Published
- 2023
- Full Text
- View/download PDF
10. Supplemental Figure S11 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S11
- Published
- 2023
- Full Text
- View/download PDF
11. Supplemental Figure S2 from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental Figure S2
- Published
- 2023
- Full Text
- View/download PDF
12. Supplemental information from Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Jing Ji, Yun Chen, Yan Qu, Yongping You, Chong Li, Liang Fan, Honglu Chao, Zhongyuan Bao, Guangchi Sun, Pengzhan Zhao, Zhenyao Chen, and Yiming Tu
- Abstract
Supplemental information
- Published
- 2023
- Full Text
- View/download PDF
13. Loss of deubiquitylase USP2 triggers development of glioblastoma via TGF-β signaling
- Author
-
Yiming Tu, Lei Xu, Jia Xu, Zhongyuan Bao, Wei Tian, Yangfan Ye, Guangchi Sun, Zong Miao, Honglu Chao, Yongping You, Ning Liu, and Jing Ji
- Subjects
Cancer Research ,Transforming Growth Factor beta ,Ubiquitination ,Genetics ,Humans ,Glioblastoma ,Polyubiquitin ,Ubiquitin Thiolesterase ,Molecular Biology ,Signal Transduction ,Smad7 Protein - Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor as one of the deadliest cancers. The TGF-β signaling acts as an oncogenic factor in GBM, and plays vital roles in development of GBM. SMAD7 is a major inhibitor of TGF-β signaling, while the deubiquitination of SMAD7 has been poorly studied in GBM. Here, we found USP2 as a new prominent candidate that could regulate SMAD7 stability. USP2 was lost in GBM, leading to the poor prognosis in patients. Moreover, aberrant DNA methylation mediated by DNMT3A induced the low expression of USP2 in GBM. USP2 depletion induced TGF-β signaling and progression of GBM. In contrast, overexpressed USP2 suppressed TGF-β signaling and GBM development. Specifically, USP2 interacted with SMAD7 and prevented SMAD7 ubiquitination. USP2 directly cleaved Lys27- and Lys48-linked poly-ubiquitin chains of SMAD7, and Lys27-linked poly-ubiquitin chains of SMAD7 K185 mediated the recruitment of SMAD7 to HERC3, which regulated Lys63-linked poly-ubiquitination of SMAD7. Moreover, we demonstrated that the DNMT3A inhibitor SGI-1027 induced USP2, suppressed TGF-β signaling and GBM development. Thus, USP2 repressed development of GBM by inhibition TGF-β signaling pathway via the deubiquitination of SMAD7.
- Published
- 2022
- Full Text
- View/download PDF
14. Endoscopic transcranial transdiaphragmatic approach in a single-stage surgery for giant pituitary adenomas
- Author
-
Xuechao Wu, Zhongyuan Bao, Wei Tian, Jing Wang, Zengli Miao, Qing Wang, and Xiaojie Lu
- Subjects
Cancer Research ,Oncology - Abstract
BackgroundThe treatment for giant pituitary adenomas (GPAs, maximal diameter >4 cm) remains challenging, with remarkable mortality and morbidity, and there is no consensus on the optimal surgical approach. Gross total resection (GTR) for GPAs is difficult to achieve through a single transsphenoidal or transcranial approach. Any residual tumor is at risk for postoperative apoplexy. In this study, we propose a new surgical technique for resecting the GPAs in a sing-stage transcranial surgery.MethodsA retrospective review of 4 patients with complicated GPAs, who had been treated via an endoscopic transcranial transdiaphragmatic approach in a single-stage surgery after routine transcranial resection, was performed. The following data was analyzed: clinical characteristics, preoperative imaging studies, resection rate, perioperative morbidity and mortality, as well as postoperative outcomes.ResultsAll patients had nonfunctioning GPAs and preoperative visual disturbances. In three patients, GTR was achieved, and in one patient, near-total resection (90%-100% of the tumor) was achieved. Three patients attained improved postoperative visual function, while one patient’s vision remained unchanged. One patient suffered a deficiency in adrenocorticotropic hormone along with thyroid-stimulating hormone, and one patient developed diabetes insipidus. Notably, none of the patients suffered cerebrospinal fluid leakage. However, one patient developed an epidural hematoma and underwent decompressive craniectomy.ConclusionsThe endoscopic transcranial transdiaphragmatic approach in a single-stage surgery can be efficiently and safely performed for maximal excision of GPAs with extensive suprasellar extension. Furthermore, relative to the conventional combined or staged approaches, this innovative surgical strategy provides neurosurgeons with a clear operative field with reduced invasiveness.
- Published
- 2023
- Full Text
- View/download PDF
15. Computed Tomography Angiography-Based Thrombus Radiomics for Predicting the Time Since Stroke Onset
- Author
-
Yue Cheng, Sunli Wan, Wenjuan Wu, Fangming Chen, Jingxuan Jiang, Dongmei Cai, Zhongyuan Bao, Yuehua Li, and Lei Zhang
- Subjects
Radiology, Nuclear Medicine and imaging - Published
- 2023
- Full Text
- View/download PDF
16. MEF2C silencing downregulates NF2 and E-cadherin and enhances Erastin-induced ferroptosis in meningioma
- Author
-
Qing Xie, Chong Li, Daijun Wang, Hiroaki Wakimoto, Lingyang Hua, Jing Ji, Gong Ye, Zhongyuan Bao, and Yangfan Ye
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Programmed cell death ,Cell ,Piperazines ,CDH1 ,Lipid peroxidation ,Mice ,chemistry.chemical_compound ,Antigens, CD ,Cell Line, Tumor ,Meningeal Neoplasms ,otorhinolaryngologic diseases ,medicine ,Animals ,Ferroptosis ,Humans ,Gene silencing ,Gene Silencing ,Neurofibromin 2 ,Gene knockdown ,biology ,MEF2 Transcription Factors ,Chemistry ,Cadherins ,Xenograft Model Antitumor Assays ,nervous system diseases ,Merlin (protein) ,Editorial ,medicine.anatomical_structure ,Oncology ,Basic and Translational Investigations ,biology.protein ,Cancer research ,Neurology (clinical) ,Meningioma ,Chromatin immunoprecipitation - Abstract
Background Ferroptosis, a programmed cell death characterized by lipid peroxidation, is implicated in various diseases including cancer. Although cell density-dependent E-cadherin and Merlin/Neurofibromin (NF2) loss can modulate ferroptosis, the role of ferroptosis and its potential link to NF2 status and E-cadherin expression in meningioma remain unknown. Methods Relationship between ferroptosis modulators expression and NF2 mutational status was examined in 35 meningiomas (10 NF2 loss and 25 NF2 wild type). The impact of NF2 and E-cadherin on ferroptosis were examined by lactate dehydrogenase (LDH) release, lipid peroxidation, and western blot assays in IOMM-Lee, CH157, and patient-derived meningioma cell models. Luciferase reporter and chromatin immunoprecipitation assays were used to assess the ability of MEF2C (myocyte enhancer factor 2C) to drive expression of NF2 and CDH1 (E-cadherin). Therapeutic efficacy of Erastin-induced ferroptosis was tested in xenograft mouse models. Results Meningioma cells with NF2 inactivation were susceptible to Erastin-induced ferroptosis. Meningioma cells grown at higher density increased expression of E-cadherin, which suppressed Erastin-induced ferroptosis. Maintaining NF2 and E-cadherin inhibited ferroptosis-related lipid peroxidation and meningioma cell death. MEF2C was found to drive the expression of both NF2 and E-cadherin. MEF2C silencing enhanced Erastin-induced ferroptotic meningioma cell death and lipid peroxidation levels in vitro, which was limited by forced expression of MEF2C targets, NF2 and E-cadherin. In vivo, anti-meningioma effect of Erastin was augmented by MEF2C knockdown and was counteracted by NF2 or E-cadherin. Conclusions NF2 loss and low E-cadherin create susceptibility to ferroptosis in meningioma. MEF2C could be a new molecular target in ferroptosis-inducing therapies for meningioma.
- Published
- 2021
- Full Text
- View/download PDF
17. DRD1 signaling enhances recovery after traumatic brain injury via an autophagy dependent pathway
- Author
-
Chao Lin, Hanxiao Chang, Zhongyuan Bao, Hui Luo, Lei Xu, Wei Wu, Ning Liu, and Jing Ji
- Abstract
Although the neurotransmitter dopamine (DA) plays a crucial pathophysiologic role after traumatic brain injury (TBI), its function and specific underlying mechanisms of action remain unclear. In this study, DA inhibits neural death induced by TBI or stretch injury, which is mediated via the dopamine receptor D1 (DRD1). Our results showed that DRD1 signaling promotes receptor interacting protein kinase 1 (RIPK1) ubiquitination via the E3 ubiquitin ligase Chip and degradation through autophagy. Importantly, in vivo data revealed that DRD1 signaling prevented neuronal death, suppressed neuroinflammation, and restored many TBI-related functional sequelae through an autophagy-dependent mechanism. These data reveal a novel mechanism involving dopamine, and suggest that DRD1 activation positively regulates Chip-mediated ubiquitylation of RIPK1—leading to its autophagic degradation.
- Published
- 2022
- Full Text
- View/download PDF
18. Distinct clinical outcome of microcystic meningioma as a WHO grade 1 meningioma subtype
- Author
-
Leihao Ren, Lingyang Hua, Zhongyuan Bao, Jiaojiao Deng, Daijun Wang, Jiawei Chen, Hong Chen, Tareq A. Juratli, Hiroaki Wakimoto, and Ye Gong
- Subjects
Cancer Research ,Neurology ,Oncology ,Neurology (clinical) - Abstract
Objective To evaluate the clinicopathological characteristics, radiology, and long-term outcomes of microcystic meningiomas (MM) and compare it with other subtypes of meningiomas managed at a single neurosurgical center. Methods A total of 87 consecutive patients who underwent surgical resection and were diagnosed as MM between 2005 to 2016 were enrolled for analysis. Clinicopathological, radiology, and prognostic information was collected and analyzed. Progression free survival (PFS) was compared with 659 patients with other subtypes of WHO grade 1 meningiomas and 167 patients with atypical meningiomas treated during the same period. Results 56 females and 31 males with MM were analyzed. Peri-tumor brain edema was frequent on T2 WI (85%).12 patients (13.8%) experienced tumor progression during the mean follow-up of 101.66 ± 40.92 months. The median PFS was unavailable, and the 5-, 10-, and 15-year progression-free rates were 96.9%, 84.0%, and 73.9%, respectively. Univariate COX analysis demonstrated skull base location, subtotal resection, and higher Ki-67 index as significant negative prognostic factors for PFS (P
- Published
- 2022
19. Up‐regulation of CHMP4B alleviates microglial necroptosis induced by traumatic brain injury
- Author
-
Binglin Chen, Jing Ji, Honglu Chao, Chong Li, Zhongyuan Bao, Pengzhan Zhao, Yiming Tu, Xiaoliu Du, Liang Fan, and Guangchi Sun
- Subjects
Adult ,Male ,0301 basic medicine ,Programmed cell death ,Traumatic brain injury ,Necroptosis ,microglia ,necroptosis ,FOXO1 ,Brain damage ,Cell Line ,Mice ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Brain Injuries, Traumatic ,Animals ,Humans ,Medicine ,CHMP4B ,Promoter Regions, Genetic ,Transcription factor ,Neuroinflammation ,Aged ,Inflammation ,Endosomal Sorting Complexes Required for Transport ,Microglia ,Forkhead Box Protein O1 ,business.industry ,traumatic brain injury ,Brain ,Original Articles ,Cell Biology ,Middle Aged ,medicine.disease ,Up-Regulation ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Original Article ,medicine.symptom ,business ,Neuroscience - Abstract
Microglial cells are key component of central nervous system (CNS) and mediate the immune response of the brain under physiological or pathological conditions. It tends to activate into a pro‐inflammatory M1 phenotype after traumatic brain injury (TBI) and promote secondary brain damage. Recently, necroptosis was found to promote microglial activation and neuroinflammation after TBI. However, the mechanism and specific interventions of microglial necroptosis after TBI remain poorly investigated. Here, we reported that overexpress the charged multivesicular body protein 4b (CHMP4B) which is a core member of the endosomal sorting required for transport complex III (ESCRT‐III) significantly decreased the level of necroptosis in microglia, improved neurological function recovery and protected against cell death after TBI. Further investigation showed that forkhead transcription factor O1 (FOXO1) was a crucial transcription factor that increased CHMP4B transcription by binding to the promoter region, thereby inhibiting necroptosis in microglia. Collectively, our findings demonstrated that CHMP4B relieved microglial necroptosis and neuroinflammation after TBI, and promote the recovery of nerve function. FOXO1 is an important factor in promoting CHMP4B expression. This study provides the novel viewpoint for TBI prevention and treatment.
- Published
- 2020
- Full Text
- View/download PDF
20. Smoothened Promotes Glioblastoma Radiation Resistance Via Activating USP3-Mediated Claspin Deubiquitination
- Author
-
Chong Li, Yiming Tu, Liang Fan, Yan Qu, Yongping You, Guangchi Sun, Zhongyuan Bao, Zhenyao Chen, Jing Ji, Pengzhan Zhao, Yun Chen, and Honglu Chao
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,DNA Repair ,Mice, Nude ,urologic and male genital diseases ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Ubiquitin ,In vivo ,Cell Line, Tumor ,Radioresistance ,Biomarkers, Tumor ,Animals ,Humans ,Radiosensitivity ,Adaptor Proteins, Signal Transducing ,Aged ,biology ,Brain Neoplasms ,urogenital system ,Chemistry ,Middle Aged ,Smoothened Receptor ,Xenograft Model Antitumor Assays ,In vitro ,nervous system diseases ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Ubiquitin-Specific Proteases ,Neoplasm Recurrence, Local ,Glioblastoma ,Smoothened ,Signal Transduction - Abstract
Purpose: Glioblastoma (GBM) is one of the most aggressive and lethal cancer types in humans. The standard treatment approach is surgery followed by chemoradiation. However, the molecular mechanisms of innate tumor radioresistance remain poorly understood. Experimental Design: We tested the expression of Smoothened (Smo) in primary and recurrent GBM tissues and cells. Then, we determined radiation effectiveness against primary and recurrent GBM cells. Lastly, the functional role of Smo in GBM radioresistance was further confirmed by in vitro and in vivo experiments. Results: We reported that Smo was significantly upregulated in recurrent GBM cell lines and tumor tissues following radiation treatment. Higher Smo expression indicated poor prognosis of GBM patients after radiation treatment. Smo had radioresistance effects in both GBM cells and human tumor xenografts. The mechanisms underlying these effects involved the attenuation of DNA damage repair caused by IR. Importantly, we found that the effect of Smo on radioresistance was mediated by Claspin polyubiquitination and proteasomal degradation, leading to the regulation of ATR–Chk1 signaling. Moreover, we found that Smo reduced Claspin polyubiquitination and proteasomal degradation by promoting USP3 transcription. Furthermore, we demonstrated that the Smo inhibitor GDC-0449 induced radiosensitivity to GBM. Conclusions: These data suggest that Smo confers radiation resistance in GBM by promoting USP3 transcription, leading to the activation of Claspin-dependent ATR–Chk1 signaling. These findings identify a potential mechanism of GBM resistance to radiation and suggest a potential therapeutic target for radiation resistance in GBM.
- Published
- 2020
- Full Text
- View/download PDF
21. Microcystic Meningioma Has A Worse Outcome Than Other WHO Grade 1 Subtypes
- Author
-
Leihao Ren, Lingyang Hua, Zhongyuan Bao, Jiaojiao Deng, Daijun Wang, Jiawei Chen, Hong Chen, Tareq A. Juratli, Hiroaki Wakimoto, and Ye Gong
- Abstract
Objective: The aim is to evaluate the clinical, radiological features and long-term outcomes of microcystic meningiomas (MM) in a single neurosurgical center.Methods: A total of 87 consecutive patients underwent surgical resection of MM between 2005 to 2016 were enrolled for analysis. Clinical, pathological, radiological and prognostic information was collected and analyzed; Univariate and multivariate COX analysis was conducted to select factors affected the progression-free survival (PFS). PFS was compared among other Grade 1 subtypes in our center as well.Results: 56 females and 31 males were identified. 12 patients (13.8%) experienced tumor progression. The median PFS was unavailable, and the 5-, 10-, and 15-year PFS rates were 96.9%, 84.0%, and 73.9%, respectively. Peri-tumor brain edema (PTBE) was frequent in MMs (85%). Univariate COX analysis demonstrated that skull base location, STR, and higher Ki-67 were significant negative prognostic factors for PFS (P < 0.05), while tumor location and Ki-67 were independent factors (P < 0.01). MM had a worse prognosis when compared to other WHO Grade 1 subtypes diagnosed in our neurosurgical center during the same time period (P = 0.0087).Conclusions: MM is a rare subtype of grade 1 meningioma, PTBE and reticular enhancement were characteristic in MMs. skull base location and higher Ki-67 labeling index were independent negative prognostic factors. MMs had a shorter PFS than the other WHO Grade 1 subtypes.
- Published
- 2022
- Full Text
- View/download PDF
22. Hsp90 Induces Acsl4-dependent Glioma Ferroptosis via Dephosphorylate Ser637 at Drp1
- Author
-
Ning Liu, Zong Miao, Wei Tian, Zhongyuan Bao, Guangchi Sun, Lei Xu, Chong Li, Yangfan Ye, Yiming Tu, honglu Chao, Sin Man Lam, and Jing Ji
- Abstract
Background: Ferroptosis is a newly identified form of regulated cell death (RCD) characterized by the iron-dependent lipid reactive oxygen species (ROS) accumulation, but its exact mechanism in gliomas remains elusive. Acyl–coenzyme A (CoA) synthetase long-chain family member 4 (Acsl4), a pivotal enzyme in the regulation of lipid biosynthesis, has been found to benefit the initiation of ferroptosis, but its role in gliomas likewise needs clarification. Erastin, widely investigated as an inducer of ferroptosis, was recently found to regulate lipid peroxidation by regulating Acsl4 other than glutathione peroxidase 4 (GPX4) in ferroptosis. Methods: Relationship between Hsp90, Drp1 and Acsl4 was determined by Co-immunoprecipitation/ Mass spectrometry and western blot assay. The impact of Hsp90 and Drp1 on Acsl4-dependent ferroptosis was examined by lipid peroxidation indicators in patient-derived PL1 and PG7 cells. The morphological changes of mitochondria are observed by confocal-fluorescence microscopy and transmission electron microscope. Therapeutic efficacy of Erastin-induced ferroptosis in vivo was examined in xenograft mouse models.Results: In this study, we demonstrated that heat shock protein 90 (Hsp90) and dynamin-related protein 1 (Drp1) actively regulated Acsl4 expression in erastin-induced ferroptosis in gliomas. Hsp90 overexpression and calcineurin (CN)–mediated Drp1 dephosphorylation at serine 637 (Ser637) promoted ferroptosis by altering mitochondrial morphology and increasing Acsl4-mediated lipid peroxidation. Importantly, the Hsp90–Acsl4 pathway mediated Acsl4-dependent ferroptosis, amplifying the anticancer activity of erastin in vitro and in vivo. Conclusions: Our study not only uncovered an important role of Hsp90–Drp1–Acsl4 pathway in erastin-induced ferroptosis but also reveals an efficient mechanism of Acsl4 as a potential therapeutic target to ferroptosis-mediated glioma therapy.
- Published
- 2022
- Full Text
- View/download PDF
23. Hsp90 induces Acsl4-dependent glioma ferroptosis via dephosphorylating Ser637 at Drp1
- Author
-
Zong Miao, Wei Tian, Yangfan Ye, Wei Gu, Zhongyuan Bao, Lei Xu, Guangchi Sun, Chong Li, Yiming Tu, Honglu Chao, Sin Man Lam, Ning Liu, and Jing Ji
- Subjects
Dynamins ,Cancer Research ,Cellular and Molecular Neuroscience ,Immunology ,Coenzyme A Ligases ,Serine ,Ferroptosis ,Humans ,Cell Biology ,Glioma ,Lipids - Abstract
Ferroptosis is a newly identified form of regulated cell death (RCD) characterized by the iron-dependent lipid reactive oxygen species (ROS) accumulation, but its mechanism in gliomas remains elusive. Acyl–coenzyme A (CoA) synthetase long-chain family member 4 (Acsl4), a pivotal enzyme in the regulation of lipid biosynthesis, benefits the initiation of ferroptosis, but its role in gliomas needs further clarification. Erastin, a classic inducer of ferroptosis, has recently been found to regulate lipid peroxidation by regulating Acsl4 other than glutathione peroxidase 4 (GPX4) in ferroptosis. In this study, we demonstrated that heat shock protein 90 (Hsp90) and dynamin-related protein 1 (Drp1) actively regulated and stabilized Acsl4 expression in erastin-induced ferroptosis in gliomas. Hsp90 overexpression and calcineurin (CN)–mediated Drp1 dephosphorylation at serine 637 (Ser637) promoted ferroptosis by altering mitochondrial morphology and increasing Acsl4-mediated lipid peroxidation. Importantly, promotion of the Hsp90–Acsl4 pathway augmented anticancer activity of erastin in vitro and in vivo. Our discovery reveals a novel and efficient approach to ferroptosis-mediated glioma therapy.
- Published
- 2021
24. Fetuin-A Alleviates Neuroinflammation Against Traumatic Brain Injury-Induced Microglial Necroptosis By Regulating Nrf-2/HO-1 Pathway
- Author
-
Jing Ji, Chong Li, Binglin Chen, Pengzhan Zhao, Guangchi Sun, Honglu Chao, Yiming Tu, and Zhongyuan Bao
- Subjects
business.industry ,Traumatic brain injury ,Necroptosis ,Medicine ,business ,medicine.disease ,Neuroscience ,Fetuin ,Neuroinflammation - Abstract
Background: The microglia-mediated inflammatory response is a vital mechanism of secondary damage following traumatic brain injury (TBI), but its underlying mechanism of microglial activation is unclear. Methods: Controlled cortical impact (CCI) was induced in adult male C57BL/6J mice, and we also used glutamate to construct a classical in vitro injury model in BV2 cell line. The activation of microglia was determined by western blot assessments and immunostaining. The inflammatory factors were determined by ELLSA. The oxidative stress marker and mitochondrial ROS were determined by immunoblotting and MitoSox Red staining. Transmission electron microscopy (TEM) was used to observe a typical morphology of necroptotic cells. Results: Our quantitative proteomics identified 2499 proteins, 157 were significantly differentially expressed between brain tissues at 6 hours after CCI (CCI6h) and sham groups, and 109 were significantly differentially expressed between CCI24h and sham brain tissues. Moreover, compared with sham groups, the terms “acute-phase response”, “inflammation”, and “protein binding” were significantly enriched in CCI groups. Interestingly, fetuin-A, a liver-secreted acute-phase glycoprotein, was involved in these biological processes. Using experimental TBI models, we found that the fetuin-A level peaked at 6 h and then decreased gradually. Importantly, we showed that fetuin-A reduced the cortical lesion volume and edema area and inhibited the inflammatory response, which was associated with suppressing microglial necroptosis, thus decreasing microglial polarization to the M1 phenotype. Furthermore, administration of fetuin-A attenuated mitochondrial oxidative stress in glutamate-treated BV2 cells, which is a critical mechanism of necroptosis suppression. In addition, we demonstrated that fetuin-A treatment promoted translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) from the cytoplasm to the nucleus in vivo; however, the Nrf-2 inhibitor ML385 and si-heme oxygenase-1 (HO-1) disrupted the regulation of oxidative stress by fetuin-A and induced increased ROS levels and necroptosis in glutamate-treated BV2 cells. Interestingly, the mechanism of fetuin-A in BV2 cells also protects neurons from adverse factors in co-culture assays.Conclusions: Our results demonstrate that fetuin-A activates Nrf-2/HO-1, suppresses oxidative stress and necroptosis levels, and thereby attenuates the abnormal inflammatory response following TBI, providing a potential therapeutic strategy for TBI treatment.
- Published
- 2021
- Full Text
- View/download PDF
25. Prokineticin-2 prevents neuronal cell deaths in a model of traumatic brain injury
- Author
-
Chong Li, Honglu Chao, Yiming Tu, Guangchi Sun, Xiupeng Xu, Hülya Bayır, Yinlong Liu, Yangfan Ye, Zhongyuan Bao, Jing Ji, Pengzhan Zhao, Zong Miao, Ning Liu, Sin Man Lam, Binglin Chen, Valerian E. Kagan, Xiaoming Wang, and Yan Liu
- Subjects
0301 basic medicine ,Male ,Ubiquitylation ,Cell ,General Physics and Astronomy ,Brain injuries ,Pathogenesis ,Lipid peroxidation ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Ubiquitin ,Brain Injuries, Traumatic ,Cells, Cultured ,Phospholipids ,Cerebral Cortex ,Neurons ,Gene knockdown ,Multidisciplinary ,biology ,Middle Aged ,Cell biology ,Mitochondria ,medicine.anatomical_structure ,Gene Knockdown Techniques ,Female ,Adult ,Programmed cell death ,Traumatic brain injury ,Science ,Primary Cell Culture ,ACSL4 ,General Biochemistry, Genetics and Molecular Biology ,Article ,Gastrointestinal Hormones ,03 medical and health sciences ,Coenzyme A Ligases ,medicine ,Animals ,Ferroptosis ,Humans ,Regeneration and repair in the nervous system ,Aged ,business.industry ,F-Box Proteins ,Neuropeptides ,Ubiquitination ,General Chemistry ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Proteolysis ,biology.protein ,Lipid Peroxidation ,business ,030217 neurology & neurosurgery - Abstract
Prokineticin-2 (Prok2) is an important secreted protein likely involved in the pathogenesis of several acute and chronic neurological diseases through currently unidentified regulatory mechanisms. The initial mechanical injury of neurons by traumatic brain injury triggers multiple secondary responses including various cell death programs. One of these is ferroptosis, which is associated with dysregulation of iron and thiols and culminates in fatal lipid peroxidation. Here, we explore the regulatory role of Prok2 in neuronal ferroptosis in vitro and in vivo. We show that Prok2 prevents neuronal cell death by suppressing the biosynthesis of lipid peroxidation substrates, arachidonic acid-phospholipids, via accelerated F-box only protein 10 (Fbxo10)-driven ubiquitination, degradation of long-chain-fatty-acid-CoA ligase 4 (Acsl4), and inhibition of lipid peroxidation. Mice injected with adeno-associated virus-Prok2 before controlled cortical impact injury show reduced neuronal degeneration and improved motor and cognitive functions, which could be inhibited by Fbxo10 knockdown. Our study shows that Prok2 mediates neuronal cell deaths in traumatic brain injury via ferroptosis., Prokineticin-2 (Prok2) is a secreted protein involved in many physiological processes. Here, the authors show that Prok2 prevents neuronal cell ferroptosis after traumatic brain injury and its administration before cortical injury reduces neuronal degeneration, and motor and cognitive impairments.
- Published
- 2021
26. S100A11 functions as novel oncogene in glioblastoma via S100A11/ANXA2/NF‐κB positive feedback loop
- Author
-
Liang Fan, Honglu Chao, Guangchi Sun, Jing Ji, Yiming Tu, Chong Li, Ailiang Zeng, Zhongyuan Bao, Xiaoliu Du, Minhong Pan, Peng Xie, and Pengzhan Zhao
- Subjects
Male ,0301 basic medicine ,Transcription, Genetic ,Carcinogenesis ,urologic and male genital diseases ,chemistry.chemical_compound ,0302 clinical medicine ,Ubiquitin ,Cell Movement ,Annexin A2 ,Feedback, Physiological ,Mice, Inbred BALB C ,Gene knockdown ,biology ,Brain Neoplasms ,Protein Stability ,S100 Proteins ,NF-kappa B ,Prognosis ,female genital diseases and pregnancy complications ,Hedgehog signaling pathway ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Molecular Medicine ,Original Article ,Stem cell ,Signal Transduction ,Proteasome Endopeptidase Complex ,Epithelial-Mesenchymal Transition ,Mice, Nude ,ubiquitination ,03 medical and health sciences ,ANXA2 ,Cell Line, Tumor ,Spheroids, Cellular ,Glioma ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Cell Proliferation ,Oncogene ,urogenital system ,Cell growth ,NF‐κB ,glioblastoma ,NF-κB ,Original Articles ,Oncogenes ,Cell Biology ,medicine.disease ,nervous system diseases ,030104 developmental biology ,chemistry ,S100A11 ,Proteolysis ,biology.protein ,Cancer research - Abstract
Glioblastoma (GBM) is the most universal type of primary brain malignant tumour, and the prognosis of patients with GBM is poor. S100A11 plays an essential role in tumour. However, the role and molecular mechanism of S100A11 in GBM are not clear. Here, we found that S100A11 was up‐regulated in GBM tissues and higher S100A11 expression indicated poor prognosis of GBM patients. Overexpression of S100A11 promoted GBM cell growth, epithelial‐mesenchymal transition (EMT), migration, invasion and generation of glioma stem cells (GSCs), whereas its knockdown inhibited these activities. More importantly, S100A11 interacted with ANXA2 and regulated NF‐κB signalling pathway through decreasing ubiquitination and degradation of ANXA2. Additionally, NF‐κB regulated S100A11 at transcriptional level as a positive feedback. We also demonstrated the S100A11 on tumour growth in GBM using an orthotopic tumour xenografting. These data demonstrate that S100A11/ANXA2/NF‐κB positive feedback loop in GBM cells that promote the progression of GBM.
- Published
- 2019
- Full Text
- View/download PDF
27. Nuclear receptor coactivator 4-mediated ferritinophagy contributes to cerebral ischemia-induced ferroptosis in ischemic stroke
- Author
-
Guangchi Sun, Zong Miao, Lin Zhao, Chong Li, Yangfan Ye, Honglu Chao, Jinyan He, Jingming Hu, Binglin Chen, Yongping You, Ning Liu, Zhongyuan Bao, Lei Xu, Pengzhan Zhao, and Jing Ji
- Subjects
Male ,Programmed cell death ,Pyrrolidines ,Nuclear Receptor Coactivators ,Ischemia ,Neuroprotection ,Downregulation and upregulation ,Malondialdehyde ,Coactivator ,Medicine ,Animals ,Ferroptosis ,Pyrroles ,Stroke ,Cells, Cultured ,Ischemic Stroke ,Pharmacology ,Neurons ,Glutathione Peroxidase ,business.industry ,Autophagy ,Brain ,Infarction, Middle Cerebral Artery ,medicine.disease ,Glutathione ,Mice, Inbred C57BL ,Nuclear receptor ,Reperfusion Injury ,Cancer research ,Lipid Peroxidation ,business ,Ubiquitin Thiolesterase - Abstract
Ischemic stroke poses a significant health risk due to its high rate of disability and mortality. To address this problem, several therapeutic approaches have been proposed, including interruption targeting programmed cell death (PCD). Ferroptosis is a newly defined PCD characterized by iron-dependent accumulation of lipid peroxidation, and is becoming a promising target for treating numerous diseases. To explore the underlying mechanisms of the initiation and execution of ferroptosis in ischemic stroke, we established stroke models in vivo and in vitro simulating ischemia/reperfusion (I/R) neuronal injury. Different from previous reports on stroke, we tested ferroptosis by measuring the levels of core proteins, such as ACSL4, 15-LOX2, Ferritin and GPX4. In addition, I/R injury induces excessive degradation of ferritin via the autophagy pathway and subsequent increase of free iron in neurons. This phenomenon has recently been termed ferritinophagy and reported to be regulated by nuclear receptor coactivator 4 (NCOA4) in some cell lines. Increased NCOA4 in cytoplasm was detected in our study and then silenced by shRNA to investigate its function. Both in vivo and in vitro, NCOA4 deletion notably abrogated ferritinophagy caused by I/R injury and thus inhibited ferroptosis. Furthermore, we found that NCOA4 was upregulated by ubiquitin specific peptidase 14 (USP14) via a deubiquitination process in damaged neurons, and we found evidence of pharmacological inhibition of USP14 effectively reducing NCOA4 levels to protect neurons from ferritinophagy-mediated ferroptosis. These findings suggest a novel and effective target for treating ischemic stroke.
- Published
- 2021
28. Hsp90 Induces Acsl4-Dependent Ferroptosis Via Dephosphorylate Ser637 at Drp1 in Glioma Cells
- Author
-
Zong Miao, Wei Tian, Zhongyuan Bao, Guangchi Sun, Lei Xu, Chong Li, Yangfan Ye, Yiming Tu, Honglu Chao, Sin Man Lam, Ning Liu, and Jing Ji
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2021
- Full Text
- View/download PDF
29. Human Cerebral Organoid Implantation Alleviated the Neurological Deficits of Traumatic Brain Injury in Mice
- Author
-
Jing Ji, Yan Liu, Zhongyuan Bao, Chen Zhang, Chaojuan Yang, Kaiheng Fang, Chong Li, Qiang Yu, Zong Miao, and Zengli Miao
- Subjects
Male ,Aging ,Pathology ,medicine.medical_specialty ,Article Subject ,Traumatic brain injury ,Mice, SCID ,Transfection ,Biochemistry ,Glial scar ,Lesion ,Mice ,Organoid ,Medicine ,Animals ,Humans ,Cerebral Cortex ,QH573-671 ,business.industry ,Cell Biology ,General Medicine ,Cortical neurons ,medicine.disease ,Embryonic stem cell ,Organoids ,Disease Models, Animal ,Spatial learning ,medicine.symptom ,business ,Cytology ,Cerebral organoid ,Research Article - Abstract
Traumatic brain injury (TBI) causes a high rate of mortality and disability, and its treatment is still limited. Loss of neurons in damaged area is hardly rescued by relative molecular therapies. Based on its disease characteristics, we transplanted human embryonic stem cell- (hESC-) derived cerebral organoids in the brain lesions of controlled cortical impact- (CCI-) modeled severe combined immunodeficient (SCID) mice. Grafted organoids survived and differentiated in CCI-induced lesion pools in mouse cortical tissue. Implanted cerebral organoids differentiated into various types of neuronal cells, extended long projections, and showed spontaneous action, as indicated by electromyographic activity in the grafts. Induced vascularization and reduced glial scar were also found after organoid implantation, suggesting grafting could improve local situation and promote neural repair. More importantly, the CCI mice’s spatial learning and memory improved after organoid grafting. These findings suggest that cerebral organoid implanted in lesion sites differentiates into cortical neurons, forms long projections, and reverses deficits in spatial learning and memory, a potential therapeutic avenue for TBI.
- Published
- 2021
- Full Text
- View/download PDF
30. MMP-9 Inhibitor GM6001 Prevents the Development of ssTBI-Induced Parkinson's Disease via the Autophagy Pathway
- Author
-
Lin Zhao, Hua Lu, Wentao Li, Zheng Li, Ning Cai, Jing Ji, Ning Liu, Zhongyuan Bao, Yingyi Wang, Lijun Hou, Wei Wu, Tianwei Guo, Yongping You, and Chao Lin
- Subjects
0301 basic medicine ,Male ,Parkinson's disease ,Mice, Transgenic ,Mitochondrion ,Pharmacology ,Matrix Metalloproteinase Inhibitors ,Blood–brain barrier ,Neuroprotection ,GM6001 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Parkinsonian Disorders ,Mitophagy ,Brain Injuries, Traumatic ,Autophagy ,Medicine ,Animals ,Maze Learning ,Trauma Severity Indices ,business.industry ,Brain ,Cell Biology ,General Medicine ,Dipeptides ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Matrix Metalloproteinase 9 ,Knockout mouse ,Female ,business ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Concussion is a widely recognized environmental risk factor for neurodegenerative diseases, including Parkinson's disease (PD). Small-vessel disease of the brain has been reported to contribute to neurodegenerative diseases. In this study, we observed BBB disruption in wild-type (WT) mice, but not in matrix metalloproteinase 9 (MMP-9) knockout mice, subjected to single severe traumatic brain injury (ssTBI). Furthermore, treating ssTBI mice with the MMP-9 inhibitor GM6001 effectively maintained BBB integrity, promoted the elimination of damaged mitochondria via mitophagy, and then prevented neuronal death and progressive neurodegeneration. However, we did not observe this neuroprotective effect of MMP-9 inhibition in beclin-1-/+ mice. Collectively, these findings revealed that concussion led to BBB disruption via MMP-9, and that GM6001 prevented the development of PD via the autophagy pathway.
- Published
- 2020
31. Activation of bradykinin B2 receptor induced the inflammatory responses of cytosolic phospholipase A2 after the early traumatic brain injury
- Author
-
Zheng Li, Ning Liu, Xiupeng Xu, Yongping You, Honglu Chao, Zhongyuan Bao, Liang Fan, Chao Lin, Chao Tao, Xiaoming Wang, Yinlong Liu, Yan Liu, Lin Zhao, and Jing Ji
- Subjects
0301 basic medicine ,biology ,Chemistry ,Bradykinin ,Inflammation ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Phospholipase A2 ,medicine ,biology.protein ,Molecular Medicine ,medicine.symptom ,B2 Bradykinin Receptor ,Neuron death ,Receptor ,Molecular Biology ,Rottlerin ,030217 neurology & neurosurgery ,Protein kinase C - Abstract
Phospholipase A2 is a known aggravator of inflammation and deteriorates neurological outcomes after traumatic brain injury (TBI), however the exact inflammatory mechanisms remain unknown. This study investigated the role of bradykinin and its receptor, which are known initial mediators within inflammation activation, as well as the mechanisms of the cytosolic phospholipase A2 (cPLA2)-related inflammatory responses after TBI. We found that cPLA2 and bradykinin B2 receptor were upregulated after a TBI. Rats treated with the bradykinin B2 receptor inhibitor LF 16-0687 exhibited significantly less cPLA2 expression and related inflammatory responses in the brain cortex after sustaining a controlled cortical impact (CCI) injury. Both the cPLA2 inhibitor and the LF16-0687 improved CCI rat outcomes by decreasing neuron death and reducing brain edema. The following TBI model utilized both primary astrocytes and primary neurons in order to gain further understanding of the inflammation mechanisms of the B2 bradykinin receptor and the cPLA2 in the central nervous system. There was a stronger reaction from the astrocytes as well as a protective effect of LF16-0687 after the stretch injury and bradykinin treatment. The protein kinase C pathway was thought to be involved in the B2 bradykinin receptor as well as the cPLA2-related inflammatory responses. Rottlerin, a Protein Kinase C (PKC) δ inhibitor, decreased the activity of the cPLA2 activity post-injury, and LF16-0687 suppressed both the PKC pathway and the cPLA2 activity within the astrocytes. These results indicated that the bradykinin B2 receptor-mediated pathway is involved in the cPLA2-related inflammatory response from the PKC pathway.
- Published
- 2018
- Full Text
- View/download PDF
32. ERK1/2/mTOR/Stat3 pathway-mediated autophagy alleviates traumatic brain injury-induced acute lung injury
- Author
-
Zhongyuan Bao, Jing Ji, Liang Fan, Honglu Chao, Zheng Li, Yinlong Liu, Kuan Jiang, Xiupeng Xu, Ning Liu, Dong Wang, and Tongle Zhi
- Subjects
Male ,STAT3 Transcription Factor ,0301 basic medicine ,MAP Kinase Signaling System ,Acute Lung Injury ,Apoptosis ,Inflammation ,Lung injury ,Stat3 Signaling Pathway ,Rats, Sprague-Dawley ,03 medical and health sciences ,Brain Injuries, Traumatic ,Autophagy ,medicine ,Animals ,STAT3 ,Lung ,Molecular Biology ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Mitogen-Activated Protein Kinase 3 ,biology ,business.industry ,TOR Serine-Threonine Kinases ,Rats ,respiratory tract diseases ,030104 developmental biology ,biology.protein ,STAT protein ,Cancer research ,Molecular Medicine ,medicine.symptom ,business - Abstract
Acute lung injury (ALI) is one of several complications in patients with traumatic brain injury (TBI). Autophagy is a primary homeostatic process that promotes cell survival under stress. Accumulating evidence implicates autophagy in the pathogenesis of ALI under various conditions. However, the role of autophagy in TBI-induced ALI remains unknown. The aim of this study was to adjust autophagy with pharmacological agents to determine its functional significance in TBI-induced ALI. Rats were preconditioned with autophagy promoter rapamycin or inhibitor 3-methyladenine before they were challenged with TBI. Extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor U0126, mechanistic target of rapamycin (mTOR) inhibitor rapamycin, and signal transducer and activator of transcription 3 (Stat3) inhibitor S31-201 were used to test the role of ERK1/2/mTOR/Stat3 signaling pathway in regulating autophagy. Autophagy is activated in lung tissues after TBI. Enhancement of autophagy suppressed apoptosis, inflammation and oxidative stress in lung tissues, which were activated after TBI, whereas inhibition of autophagy aggravated these critical pathological changes. Autophagy also improved TBI-induced impairment in pulmonary barrier function, oxygenation function and static compliance. Furthermore, TBI-induced autophagy was mediated by ERK1/2/mTOR/Stat3 pathway, which may serve to reduce ALI and improve pulmonary barrier function, oxygenation function and static compliance. These findings are important for the prevention and treatment of TBI-induced ALI.
- Published
- 2018
- Full Text
- View/download PDF
33. Lowered iPLA2γ activity causes increased mitochondrial lipid peroxidation and mitochondrial dysfunction in a rotenone-induced model of Parkinson's disease
- Author
-
Zheng Li, Yan Liu, Ning Liu, Jing Ji, Zhongyuan Bao, Honglu Chao, Xiaoming Wang, Yongping You, Chao Lin, Xiupeng Xu, Yinlong Liu, and Xian Fu
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Parkinson's disease ,Neurodegeneration ,Rotenone ,Biology ,Mitochondrion ,medicine.disease ,medicine.disease_cause ,Lipid peroxidation ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Endocrinology ,Developmental Neuroscience ,Neurology ,chemistry ,Biochemistry ,Internal medicine ,medicine ,Inner mitochondrial membrane ,Oxidative stress ,Abnormal mitochondrial morphology - Abstract
iPLA2γ, calcium-independent phospholipase A2γ, discerningly hydrolyses glycerophospholipids to liberate free fatty acids. iPLA2γ-deficiency has been associated with abnormal mitochondrial function. More importantly, the iPLA2 family is causative proteins in mitochondrial neurodegenerative disorders such as parkinsonian disorders. However, the mechanisms by which iPLA2γ affects Parkinson's disease (PD) remain unknown. Mitochondrion stress has a key part in rotenone-induced dopaminergic neuronal degeneration. The present evaluation revealed that lowered iPLA2γ function provokes the parkinsonian phenotype and leads to the reduction of dopamine and its metabolites, lowered survival, locomotor deficiencies, and organismal hypersensitivity to rotenone-induced oxidative stress. In addition, lowered iPLA2γ function escalated the amount of mitochondrial irregularities, including mitochondrial reactive oxygen species (ROS) regeneration, reduced ATP synthesis, reduced glutathione levels, and abnormal mitochondrial morphology. Further, lowered iPLA2γ function was tightly linked with strengthened lipid peroxidation and mitochondrial membrane flaws following rotenone treatment, which can cause cytochrome c release and eventually apoptosis. These results confirmed the important role of iPLA2γ, whereby decreasing iPLA2γ activity aggravates mitochondrial degeneration to induce neurodegenerative disorders in a rotenone rat model of Parkinson's disease. These findings may be useful in the design of rational approaches for the prevention and treatment of PD-associated symptoms.
- Published
- 2018
- Full Text
- View/download PDF
34. Silencing Pre-B-cell leukemia homeobox 3 decreases the proliferation of human glioma cells in vitro and in vivo
- Author
-
Zhongyuan Bao, Ning Liu, Xiupeng Xu, Ning Cai, Jing Ji, and Yongping You
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Mice, Nude ,Apoptosis ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Glioma ,medicine ,Animals ,Data Mining ,Humans ,Gene silencing ,Cell Proliferation ,Homeodomain Proteins ,Mice, Inbred BALB C ,Gene knockdown ,Brain Neoplasms ,Cell growth ,Cell Cycle ,Cell cycle ,medicine.disease ,Up-Regulation ,030104 developmental biology ,Neurology ,Oncology ,Cell culture ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,B-cell leukemia ,Cancer research ,Neurology (clinical) ,Neoplasm Transplantation - Abstract
Among primary brain tumors, gliomas are the most common and most aggressive, with a poor prognosis and limited treatment options. Thus, it is essential to determine the mechanisms involved in glioma development to develop effective therapies for glioma patients. Pre-B-cell leukemia homeobox 3 (PBX3), a critical member of the PBX family, is frequently overexpressed in multiple human malignancies. However, the expression patterns and biological functions, as well as the involved molecular functions of PBX3 in human gliomas remain largely unknown. In this study, we demonstrate that PBX3 expression is increased in both human glioma tissues and cell lines compared with their normal counterparts. These results suggested that PBX3 might be involved in glioma progression. Thus, the role of PBX3 in glioma cell proliferation was investigated using genetic knockdown and overexpression methods. The results showed that PBX3 knockdown inhibited glioma cell proliferation and induced apoptosis, while PBX3 overexpression significantly promoted glioma cell proliferation. Mechanistically, we found that PBX3 promoted cell proliferation by modulating cell cycle progression. A xenograft LN229 model was used to confirm that PBX3 depletion decreased tumor growth in vivo. In summary, our findings reveal that PBX3 may be a potential therapeutic target in gliomas.
- Published
- 2017
- Full Text
- View/download PDF
35. Silencing of A20 Aggravates Neuronal Death and Inflammation After Traumatic Brain Injury: A Potential Trigger of Necroptosis
- Author
-
Xiupeng Xu, Yan Liu, Liang Fan, Xiaoming Wang, Lin Zhao, Honglu Chao, Jing Ji, Yongping You, Yinlong Liu, Zhongyuan Bao, and Ning Liu
- Subjects
0301 basic medicine ,Programmed cell death ,Traumatic brain injury ,Necroptosis ,necroptosis ,Inflammation ,HMGB1 ,Neuroprotection ,lcsh:RC321-571 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Molecular Biology ,Original Research ,biology ,business.industry ,traumatic brain injury ,medicine.disease ,neuronal death ,A20 ,030104 developmental biology ,inflammation ,biology.protein ,Cancer research ,Tumor necrosis factor alpha ,medicine.symptom ,Neuron death ,business ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Programmed cell death is an important biological process that plays an indispensable role in traumatic brain injury (TBI). Inhibition of necroptosis, a type of programmed cell death, is pivotal in neuroprotection and in preventing associated inflammatory responses. Our results showed that necroptosis occurred in human brain tissues after TBI. Necroptosis was also induced by controlled cortical impact (CCI) injury in a rat model of TBI and was accompanied by high translocation of high-mobility group box-1 (HMGB1) to the cytoplasm. HMGB1 was then passed through the impaired cell membrane to upregulate the receptor for advanced glycation end-products (RAGE), nuclear factor (NF)-κB, and inflammatory factors such as interleukin-6 (IL-6), interleukin-1 (IL-1β), as well as NACHT, LRR and PYD domains-containing protein 3 (NLRP3). Necroptosis was alleviated by necrostatin-1 and melatonin but not Z-VAD (a caspase inhibitor), which is consistent with the characteristic of caspase-independent signaling. This study also demonstrated that tumor necrosis factor, alpha-induced protein 3 (TNFAIP3, also known as A20) was indispensable for regulating and controlling necroptosis and inflammation after CCI. We found that a lack of A20 in a CCI model led to aggressive necroptosis and attenuated the anti-necroptotic effects of necrostatin-1 and melatonin.
- Published
- 2019
- Full Text
- View/download PDF
36. LINC00511 contributes to glioblastoma tumorigenesis and epithelial-mesenchymal transition via LINC00511/miR-524-5p/YB1/ZEB1 positive feedback loop
- Author
-
Pengzhan Zhao, Chong Li, Yiming Tu, Zhongyuan Bao, Jing Ji, Jinhao Li, Shuang Liu, Xiaoliu Du, and Minhong Pan
- Subjects
0301 basic medicine ,Male ,Transcription, Genetic ,Carcinogenesis ,medicine.disease_cause ,0302 clinical medicine ,Cell Movement ,LINC00511 ,ZEB1 ,Feedback, Physiological ,Gene knockdown ,Mice, Inbred BALB C ,Transition (genetics) ,Brain Neoplasms ,Cell Cycle ,Prognosis ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Molecular Medicine ,RNA, Long Noncoding ,Original Article ,Epithelial-Mesenchymal Transition ,Mice, Nude ,Biology ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Epigenetics ,Transcription factor ,Cell Proliferation ,epithelial‐mesenchymal transition ,Base Sequence ,Competing endogenous RNA ,glioblastoma ,RNA ,Zinc Finger E-box-Binding Homeobox 1 ,Cell Biology ,Original Articles ,MicroRNAs ,tumorigenesis ,030104 developmental biology ,HEK293 Cells ,Cancer research ,Y-Box-Binding Protein 1 - Abstract
Tumour invasion is closely related to the prognosis and recurrence of glioblastoma multiforme and partially attributes to epithelial‐mesenchymal transition. Long intergenic non‐coding RNA 00511 (LINC00511) plays a pivotal role in tumour; however, the role of LINC00511 in GBM, especially in the epigenetic molecular regulation mechanism of EMT, is still unclear. Here, we found that LINC00511 was up‐regulated in GBM tissues and relatively high LINC00511 expression predicted poorer prognosis. Moreover, ectopic LINC00511 enhanced GBM cells proliferation, EMT, migration and invasion, whereas LINC00511 knockdown had the opposite effects. Mechanistically, we confirmed that ZEB1 acted as a transcription factor for LINC00511 in GBM cells. Subsequently, we found that LINC00511 served as a competing endogenous RNA that sponged miR‐524‐5p to indirectly regulate YB1, whereas, up‐regulated YB1 promoted ZEB1 expression, which inversely facilitated LINC00511 expression. Finally, orthotopic xenograft models were performed to further demonstrate the LINC00511 on GBM tumorigenesis. This study demonstrates that a LINC00511/miR‐524‐5p/YB1/ZEB1 positive feedback loop provides potential therapeutic targets for GBM progression.
- Published
- 2019
37. Activation of bradykinin B2 receptor induced the inflammatory responses of cytosolic phospholipase A
- Author
-
Honglu, Chao, Yinlong, Liu, Chao, Lin, Xiupeng, Xu, Zheng, Li, Zhongyuan, Bao, Liang, Fan, Chao, Tao, Lin, Zhao, Yan, Liu, Xiaoming, Wang, Yongping, You, Ning, Liu, and Jing, Ji
- Subjects
Adult ,Male ,Receptor, Bradykinin B2 ,Phospholipases A2, Cytosolic ,Bradykinin ,Rats, Sprague-Dawley ,Young Adult ,Bradykinin B2 Receptor Antagonists ,Brain Injuries, Traumatic ,Animals ,Humans ,Benzopyrans ,Enzyme Inhibitors ,Aged ,Inflammation ,Epilepsy ,Acetophenones ,Brain ,Middle Aged ,Rats ,Up-Regulation ,Disease Models, Animal ,Animals, Newborn ,Astrocytes ,Quinolines ,Female - Abstract
Phospholipase A
- Published
- 2018
38. Lowered iPLA
- Author
-
Honglu, Chao, Yinlong, Liu, Xian, Fu, Xiupeng, Xu, Zhongyuan, Bao, Chao, Lin, Zheng, Li, Yan, Liu, Xiaoming, Wang, Yongping, You, Ning, Liu, and Jing, Ji
- Subjects
Male ,Membrane Potential, Mitochondrial ,Mitochondria ,Rats ,Group VI Phospholipases A2 ,Rats, Sprague-Dawley ,Disease Models, Animal ,Oxidative Stress ,Mesencephalon ,Cell Line, Tumor ,Rotenone ,Animals ,Humans ,Lipid Peroxidation ,Parkinson Disease, Secondary ,RNA, Small Interfering - Abstract
iPLA
- Published
- 2017
39. Corrigendum to 'Activation of bradykinin B2 receptor induced the inflammatory responses of cytosolic phospholipase A2 after the early traumatic brain injury.' [Biochem. Biophys. Acta Mol. Basis Dis. 1864/9 Pt B (2018) 2957–2971]
- Author
-
Honglu Chao, Zheng Li, Liang Fan, Xiaoming Wang, Yinlong Liu, Jing Ji, Chao Lin, Ning Liu, Xiupeng Xu, Lin Zhao, Yan Liu, Yongping You, and Zhongyuan Bao
- Subjects
Cytosol ,Phospholipase A2 ,biology ,Chemistry ,Traumatic brain injury ,Mole ,biology.protein ,medicine ,Molecular Medicine ,Pharmacology ,medicine.disease ,Molecular Biology ,Bradykinin B2 Receptor - Published
- 2019
- Full Text
- View/download PDF
40. Omega-3 fatty acids regulate NLRP3 inflammasome activation and prevent behavior deficits after traumatic brain injury
- Author
-
Ning Liu, Yongping You, Honglu Chao, Jing Ji, Yinlong Liu, Zhongyuan Bao, Xiaoming Wang, Zheng Li, Yan Liu, Lijun Hou, Chao Lin, and Xiupeng Xu
- Subjects
0301 basic medicine ,Scaffold protein ,Male ,Traumatic brain injury ,Inflammasomes ,Inflammation ,Pharmacology ,Inhibitory postsynaptic potential ,Benzoates ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,Free fatty acid receptor 1 ,Brain Injuries, Traumatic ,Fatty Acids, Omega-3 ,NLR Family, Pyrin Domain-Containing 3 Protein ,Medicine ,Animals ,Secretion ,Enzyme Inhibitors ,Receptor ,Brain Chemistry ,Behavior, Animal ,business.industry ,Caspase 1 ,Inflammasome ,medicine.disease ,beta-Arrestin 2 ,Mitochondria ,Rats ,030104 developmental biology ,Pyrimidines ,Neurology ,Immunology ,Cytokines ,medicine.symptom ,business ,030217 neurology & neurosurgery ,medicine.drug ,Interleukin-1 - Abstract
Omega-3 fatty acids (ω-3 FAs) attenuate inflammation and improve neurological outcome in response to traumatic brain injury (TBI), but the specific anti-inflammatory mechanisms remain to be elucidated. Here we found that NLRP3 inflammasome and subsequent pro-inflammatory cytokines were activated in human brains after TBI. Rats treated with ω-3 FAs had significantly less TBI-induced caspase-1 cleavage and IL-1β secretion than those with vehicle. G protein-coupled receptor 40 (GPR40) was observed to be involved in this anti-inflammation. GW1100, a GPR40 inhibitor, eliminated the anti-inflammatory effect of ω-3 FAs after TBI. β-Arrestin-2 (ARRB2), a downstream scaffold protein of GPR40, was activated to inhibit inflammation via directly binding with NLRP3 in the ω-3 FAs treatment group. Interestingly, we also observed that ω-3 FAs prevented NLRP3 mitochondrial localization, which was reversed by GW1100. Furthermore, ω-3 FAs markedly ameliorated neuronal death and behavioral deficits after TBI, while GW1100 significantly suppressed this effect. Collectively, these data indicate that the GPR40-mediated pathway is involved in the inhibitory effects of ω-3 FAs on TBI-induced inflammation and ARRB2 is activated to interact with NLRP3.
- Published
- 2016
41. Corrigendum to 'ERK1/2/mTOR/Stat3 pathway-mediated autophagy alleviates traumatic brain injury-induced acute lung injury' [Biochim. Biophys. Acta 1864/5PA(2018) 1663–1674]
- Author
-
Jing Ji, Zheng Li, Kuan Jiang, Liang Fan, Xiupeng Xu, Tongle Zhi, Honglu Chao, Yinlong Liu, Ning Liu, Zhongyuan Bao, and Dong Wang
- Subjects
0301 basic medicine ,biology ,business.industry ,Traumatic brain injury ,Autophagy ,Lung injury ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Molecular Medicine ,Medicine ,STAT3 ,business ,Molecular Biology ,PI3K/AKT/mTOR pathway - Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.