11 results on '"Qichun Wei"'
Search Results
2. Trimethylamine N-oxide promotes hyperoxaluria-induced calcium oxalate deposition and kidney injury by activating autophagy
- Author
-
Xiaohua Wang, Jie Guo, Fei Gao, Chun Tang, Shan Jiang, Jiong Tian, Xiaodong Fu, Weipeng Hu, Fang Dong, Andreas Patzak, Liang Zhao, En Yin Lai, Pontus B. Persson, Linlin Liu, Qichun Wei, and Xiaoqiu Ren
- Subjects
medicine.medical_specialty ,Programmed cell death ,Calcium oxalate ,Inflammation ,Trimethylamine N-oxide ,Kidney ,medicine.disease_cause ,Biochemistry ,Methylamines ,Mice ,chemistry.chemical_compound ,Physiology (medical) ,Internal medicine ,Autophagy ,medicine ,Animals ,Hyperoxaluria ,Oxalates ,Calcium Oxalate ,Kinase ,Endocrinology ,chemistry ,Apoptosis ,medicine.symptom ,Oxidative stress - Abstract
Calcium oxalate (CaOx) is the most common component of kidney stones. Oxidative stress, inflammation and autophagy-induced cell death are the major causes of CaOx crystal deposition and CaOx crystal deposition can further lead to kidney injury. Trimethylamine N-oxide (TMAO), a gut microbiota-derived metabolite, plays an important role in the pathogenesis of many diseases, such as atherosclerosis, diabetes and chronic kidney disease, but the effect of TMAO on hyperoxaluria-induced CaOx crystal deposition and kidney injury remains unknown. We hypothesize that TMAO aggravates CaOx crystal deposition via promoting CaOx-mediated cell death. C57Bl/6 mice were given high-oxalate diet as a model of hyperoxaluria. TMAO was provided via drinking water. Serum TMAO levels increased 15 days after CaOx treatment (6.30 ± 0.17 μmol/L vs. 34.65 ± 8.95 μmol/L). High-oxalate diet induced inflammation, CaOx deposition and kidney injury, which TMAO aggravated. In accordance, TMAO intensified high-oxalate diet induced oxidative stress, autophagy and apoptosis. Moreover, TMAO enhanced CaOx crystal adhesion to HK-2 cells and reduced cell viability (from 88.9 ± 1.6% to 75.0 ± 2.7%). Protein kinase R-like endoplasmic reticulum kinase (PERK) may mediate these TMAO effects, as TMAO promoted PERK phosphorylation. Consistently, PERK knockdown alleviated TMAO-evoked CaOx-autophagy, apoptosis and oxidative stress in HK-2 cells. In conclusion, TMAO can aggravate hyperoxaluria-induced kidney injury by triggering the PERK/ROS pathway, which enhances autophagy, apoptosis and inflammation, and facilitates CaOx crystal deposition in renal tubular cells.
- Published
- 2022
3. Sulfonic acid functionalized β zeolite as efficient bifunctional solid acid catalysts for the synthesis of 5-hydroxymethylfurfural from cellulose
- Author
-
Xinyi Xing, Xian Shi, Mengya Ruan, Qichun Wei, Ying Guan, Hui Gao, and Siquan Xu
- Subjects
Structural Biology ,General Medicine ,Molecular Biology ,Biochemistry - Published
- 2023
4. Identification of a novel Immune-Related prognostic model for patients with colorectal cancer based on 3 subtypes
- Author
-
Xi Yang and Qichun Wei
- Subjects
Immunology ,Immunology and Allergy ,Hematology - Published
- 2023
5. Cisplatin and Fluorouracil Induction Chemotherapy With or Without Docetaxel in Locoregionally Advanced Nasopharyngeal Carcinoma
- Author
-
Xiaozhong Chen, Ting Jin, Wen-feng Li, Wei-feng Qin, Qichun Wei, Qi-Feng Jin, Feng Jiang, Xiao-nan Sun, and Yong-shi Jia
- Subjects
0301 basic medicine ,Oncology ,Original article ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Neutropenia ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Cisplatin ,business.industry ,Induction chemotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Radiation therapy ,030104 developmental biology ,Docetaxel ,Nasopharyngeal carcinoma ,Fluorouracil ,030220 oncology & carcinogenesis ,business ,Chemoradiotherapy ,medicine.drug - Abstract
In this study, we aim to compare the progression-free survival (PFS) rates and side effects of induction chemotherapy based on docetaxel, cisplatin and fluorouracil (TPF) versus cisplatin and fluorouracil (PF) in patients with locoregionally-advanced nasopharyngeal carcinoma who received subsequent chemoradiotherapy. We randomly assigned 278 patients with stage III or IV NPC (without distant metastases) to receive either TPF or PF induction chemotherapy, followed by cisplatin-based chemoradiotherapy every 3 weeks and intensity-modulated radiation therapy for 5 days per week. After a minimum of 2 years follow-up, a PFS benefit was observed for TPF compared to PF, though this difference was not statistically significant (84.5% vs. 77.9%, P = .380). Due to increased frequencies of grade 3 or 4 neutropenia and diarrhea, significantly more patients in the TPF group required treatment delays and dose modifications. Our findings suggest that PF induction chemotherapy has substantially better tolerance and compliance rates than TPF induction chemotherapy. However, the treatment efficacy of PF is not superior to TPF induction chemotherapy in patients with locoregionally-advanced NPC (ClinicalTrials.gov number, NCT01536223 ).
- Published
- 2019
6. Effectiveness of different treatment strategies in elderly patients with glioblastoma: An evidence map of randomized controlled trials
- Author
-
Wenbin, Ma, Xiaofang, Sheng, Guang, Li, Qichun, Wei, Zhirui, Zhou, and Xiaoguang, Qiu
- Subjects
Oncology ,Brain Neoplasms ,Humans ,Chemoradiotherapy ,Immunotherapy ,Hematology ,Middle Aged ,Neoplasm Recurrence, Local ,Glioblastoma ,Combined Modality Therapy ,Aged ,Randomized Controlled Trials as Topic - Abstract
To summarize randomized controlled trials (RCTs) evidence for the effectiveness of available treatment strategies for elderly patients with glioblastoma (GBM) (defined as 60 years old and above) and to identify research gaps, we conducted this evidence map. Finally, 22 RCTs (with 3052 participants) were included. For newly diagnosed elderly patients with GBM (16 RCTs), 75% was identified about the effectiveness of chemotherapy, radiotherapy and chemo-radiotherapy, while there was relatively less evidence evaluating targeted therapy (12.5%), immunotherapy (18.75%) and tumor treating fields (TTFs) therapy (6.25%). Less evidence was identified in elderly patients with recurrent GBM (6 RCTs), including 2 RCTs for immunotherapy and 4 RCTs for targeted therapy. Current RCTs revealed some beneficial treatment strategies. However, more are needed to optimize regimens of RT and chemo-radiotherapy, explore potential new therapies such as targeted therapy, immunotherapy and combination therapies, and identify biomarkers to guide appropriate patient and treatment selection.
- Published
- 2022
7. Response to 'Radiation induced lymphopenia in head and neck cancer: The importance of rigorous statistical analysis, radiation field size, and treatment modality'
- Author
-
Dongjun, Dai, Qiaoying, Tian, Yongjie, Shui, Jinfan, Li, and Qichun, Wei
- Subjects
Oncology ,Head and Neck Neoplasms ,Lymphopenia ,Humans ,Radiology, Nuclear Medicine and imaging ,Hematology - Published
- 2022
8. Targeting tumor hypoxia with stimulus-responsive nanocarriers in overcoming drug resistance and monitoring anticancer efficacy
- Author
-
Ming-Yi Huang-Fu, Meng-Ting Lin, Qichun Wei, Hui-Na Liu, Jian-Qing Gao, Ning-Ning Guo, Min Han, Wenhong Xu, Zhi-Qi Xie, Wang Tiantian, Wang-Wei Guo, and Chen Jiejian
- Subjects
Biomedical Engineering ,Mice, Nude ,02 engineering and technology ,Biochemistry ,Biomaterials ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Humans ,Doxorubicin ,RNA, Small Interfering ,Molecular Biology ,Zebrafish ,Sensitization ,Tumor microenvironment ,Tumor hypoxia ,Chemistry ,Neoplasms, Experimental ,General Medicine ,Hypoxia (medical) ,021001 nanoscience & nanotechnology ,Xenograft Model Antitumor Assays ,Cell Hypoxia ,medicine.anatomical_structure ,A549 Cells ,030220 oncology & carcinogenesis ,Drug delivery ,MCF-7 Cells ,Cancer research ,Nanoparticles ,Female ,Drug Monitoring ,Nanocarriers ,medicine.symptom ,Reactive Oxygen Species ,0210 nano-technology ,Biotechnology ,medicine.drug - Abstract
Although existing nanomedicines have focused on the tumor microenvironment with the goal of improving the effectiveness of conventional chemotherapy, the penetration of a tumor’s core still represents a formidable barrier for existing drug delivery systems. Therefore, a novel multifunctional hypoxia-induced size-shrinkable nanoparticle has been designed to increase the penetration of drugs, nucleic acids, or probes into tumors. This cooperative strategy relies on three aspects: (i) the responsiveness of nanoparticles to hypoxia, which shrink when triggered by low oxygen concentrations; (ii) the core of a nanoparticle involves an internal cavity and strong positive charges on the surface to deliver both doxorubicin and siRNA; and (iii) a reactive oxygen species (ROS) probe is incorporated in the nanoparticle to monitor its preliminary therapeutic response in real time, which is expected to realize the enhanced efficacy together with the ability to self-monitor the anticancer activity. A more effective inhibition of tumor growth was observed in tumor-bearing zebrafish, demonstrating the feasibility of this cooperative strategy for in vivo applications. This research highlights a promising value in delivering drugs, nucleic acids, or probes to a tumor’s core for cancer imaging and treatment. Statement of Significance Hypoxia-induced chemoresistance of tumor cells still represents a formidable barrier, as it is difficult for existing drug delivery systems to penetrate the tumor hypoxia core. This study involves the hypoxia-responsive size-shrinkable nanoparticle co-delivery of DOX and siRNA to enhance the penetration of DOX deep within tumors and subsequently disturb crucial pathways of cancer development induced by hypoxia and to improve sensitization to DOX chemotherapy. Furthermore, the nanopreparation can combine the ROS probe as a self-reporting nanopreparation to realize the function of real-time feedback efficacy, which has a good application prospect in the diagnosis and treatment of cancer.
- Published
- 2018
9. Parthanatos and its associated components: Promising therapeutic targets for cancer
- Author
-
Yali Wang, Anwen Shao, Yihan Yao, Yongchuan Deng, Sifeng Tao, Lihong Liu, Yunxiang Zhou, and Qichun Wei
- Subjects
0301 basic medicine ,Pharmacology ,PARG ,Programmed cell death ,Carcinogenesis ,medicine.medical_treatment ,Autophagy ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Targeted therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Neoplasms ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,medicine ,Animals ,Humans ,Signal transduction ,Parthanatos - Abstract
Parthanatos is a PARP1-dependent, caspase-independent, cell-death pathway that is distinct from apoptosis, necrosis, or other known forms of cell death. Parthanatos is a multistep pathway that plays a pivotal role in tumorigenesis. There are many molecules in the parthanatos cascade that can be exploited to create therapeutic interventions for cancer management, including PARP1, PARG, ARH3, AIF, and MIF. These critical molecules are involved in tumor cell proliferation, progression, invasion, and metastasis. Therefore, these molecular signals in the parthanatos cascade represent promising therapeutic targets for cancer therapy. In addition, intimate interactions occur between parthanatos and other forms of cancer cell death, such as apoptosis and autophagy. Thus, co-targeting a combination of parthanatos and other death pathways may further provide a new avenue for cancer precision treatment. In this review, we elaborate on the signaling pathways of canonical parthanatos and briefly introduce the non-canonical parthanatos. We also shed light on the role parthanatos and its associated components play in tumorigenesis, particularly with respect to the aforementioned five molecules, and discuss the promise targeted therapy of parthanatos and its associated components holds for cancer therapy.
- Published
- 2021
10. MicroRNA-34a induces a senescence-like change via the down-regulation of SIRT1 and up-regulation of p53 protein in human esophageal squamous cancer cells with a wild-type p53 gene background
- Author
-
Qichun Wei, Yuezhen Wang, Shujun Dai, Zhimin Ye, Jun Fang, Pintong Huang, Wei Feng, and Zhenfu Fu
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,0301 basic medicine ,Cancer Research ,Esophageal Neoplasms ,Down-Regulation ,Apoptosis ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Sirtuin 1 ,Cell Line, Tumor ,microRNA ,Humans ,Cytotoxic T cell ,Cellular Senescence ,Cell Proliferation ,Cell growth ,Genes, p53 ,Molecular biology ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Reverse transcription polymerase chain reaction ,MicroRNAs ,030104 developmental biology ,Oncology ,Doxorubicin ,MicroRNA 34a ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Carcinoma, Squamous Cell ,Cancer research ,Esophageal Squamous Cell Carcinoma ,Tumor Suppressor Protein p53 ,Cell aging - Abstract
MiR-34a has been reported as a non-coding RNA universally expressed in normal old cells and a probable suppressor of diverse cancer cells; however, this miRNA's expression and anti-tumor mechanism in esophageal squamous cancer cells (ESCC) remains unclear. We explored these questions in three human ESCC lines, KYSE-450, KYSE-410, and ECa-109, with wild-type p53 and mutant p53 backgrounds. Through a specific stem-loop RT primer for miR-34a, we examined the relevant expression level of miR-34a in these three cell lines using real-time reverse transcription PCR (qRT-PCR). We found that the expression level of miR-34a induced by the DNA damage agent adrmycin (ADR) was both p53- and time-dependent. Following incubation with miR-34a, cellular growth inhibition was exhibited differently in the three cell lines harbored with different p53 backgrounds. Furthermore, the MTT assay demonstrated an miR-34a-related cytotoxic effect in cell growth. Senescence-associated β-galactosidase (SA-β-Gal) staining was used to examine senescence-like phenotypes induced by miR-34a. Mechanistic investigation suggested that the down-regulation of Sirtuin1 (SIRT1) and up-regulation of p53/p21 contributed to the anti-tumor mechanism of miR-34a in wild-type p53 ECa-109 cells, while neither of the apoptosis-related proteins PARP and caspase-3 caused significant changes. In summary, our findings indicated that the intrinsic expression of miR-34a was relatively low and was expressed differently among different p53 backgrounds and ADR treatment times. The anti-tumor effect of miR-34a was primarily dependent on the regulation of SIRT1 and p53/p21 protein, not apoptosis-associated proteins.
- Published
- 2016
11. Systematic Evaluation of Paired Primary and Recurrent Gliomas Identifies Key Genetic Markers for Survival and Therapeutic Targets
- Author
-
Xiang-Long Li, Qichun Wei, Jinghong Xu, Xiaoqiu Ren, Juan Zhou, Yongjie Shu, Jing Xu, Bicheng Zhang, Yinglu Guo, Zhanhuai Wang, Wei Yu, Xiao-fang Yu, Yanqin Huang, and Zexin Chen
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Therapy group ,Disease progression ,Ethics committee ,Recurrent Glioma ,medicine.disease ,Genetic marker ,Internal medicine ,Glioma ,medicine ,Adjuvant therapy ,Immunohistochemistry ,business ,neoplasms - Abstract
Background: The differences in genetic characteristics between primary and recurrent gliomas have been poorly characterized. We investigated whether genetic characteristics in recurrent gliomas would differ from those of primary gliomas, as a means of identifying unique markers in recurrent glioma to permit a better understanding of disease progression and therapeutic considerations. Methods: Multiple genes involved in eight important pathways in primary and their corresponding paired recurrent glioma samples from 42 patients were analyzed by immunohistochemical staining. Findings: In recurrent glioma, we saw a significantly increased expression of PD-1, PD-L1, VEGF, VEGFR2, CD133, MGMT, and Rad51 when compared with their corresponding primary tumors. VEGF expression was correlated with VEGFR2 expression in both primary and recurrent gliomas, and their co-expression was associated with shorter progression-free survival (PFS) and overall survival (OS). PD-1 expression was correlated with PD-L1 expression in recurrent gliomas. As compared to primary gliomas, the overall expression of MGMT was increased in the adjuvant therapy group but was not statistically different from the no-adjuvant therapy group. The group in which MGMT increased had a shorter PFS and OS than did the group in which MGMT was not increased. Co-expression of P53 and Ki-67 in recurrent gliomas was associated with poor median OS after second resection. Interpretation: The gene expression characteristics of recurrent gliomas were significantly different from those of their corresponding primary gliomas, a result that has important implications for patient survival. Treatment based on these newly identified changes may offer better therapeutic options for recurrent glioma. Funding Statement: This work was surported by the National Natural Science Foundation of China (81572952). Declaration of Interests: The authors declare that they have no conflict of interest. Ethics Approval Statement: This study was approved by the Ethics Committee of SAHZU, and was carried out in accordance with the Declaration of Helsinki.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.