1
|
Guo C, Liu Y, Ma F, Xu X, Zhang W, Zhao Z, Wang Y, Kong Q. Microenvironment Remodeling Microgel Repairs Degenerated Intervertebral Disc via Programmed Delivery of MicroRNA-155. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 39804788 DOI: 10.1021/acsami.4c18801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The progression of intervertebral disc degeneration (IVDD) is associated with increased cell apoptosis and reduced extracellular matrix (ECM) production, both of which are driven by ongoing inflammation. Thus, alleviating the acidic inflammatory microenvironment and mitigating the apoptosis of nucleus pulposus cells (NPCs) are essential for intervertebral disc (IVD) regeneration. Regulating pH levels in the local environment can reduce inflammation and promote tissue recovery. In this study, a lactic acid-capturing microgel carrying a functionalized miRNA-155 nanocarrier was designed for IVD regeneration. microRNA-155 was loaded into the NPC-targeted nanogel via host-guest binding. The miR-155 nanocarrier (NGM) achieved lactic acid-sensitive release of miRNA-155, resulting in rapid regulation of apoptosis. Moreover, SS31, which dissociated from the nanogel network, had the ability to regulate mitochondrial metabolism. Moreover, the microgel was constructed using a matrix metalloproteinase-responsive peptide. The chitosan coating on the microgel system was ingeniously employed to capture lactic acid and enable pH-responsive dissociation, thereby alleviating the acidic microenvironment to protect cell viability and facilitate the delivery of the NGM. The microgel system effectively promoted IVD regeneration by alleviating the acidic microenvironment and preventing NPC apoptosis.
Collapse
Affiliation(s)
- Chuan Guo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuheng Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fei Ma
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xueyuan Xu
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weifei Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhen Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yu Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qingquan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
2
|
Zai H, Wu X, Zhou Y, Hu Y, Zhu Q. Lnc NBAT1 Inhibits the Proliferation and Migration of Liver Cancer Cells Through the miR-21/PDCD4/AP-1 Signaling Axis. Appl Biochem Biotechnol 2025; 197:1-18. [PMID: 39093348 DOI: 10.1007/s12010-024-05008-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
Long non-coding RNAs (Lnc RNAs) are proven to participate in liver cancer (LC) regulation. The regulation of miR-21 by lnc NBAT1 has been studied in other cancers. However, the effect of this regulation on LC and its specific mechanism remains unclear. Lnc NBAT1 and miR-21 expressions in clinical tissues were measured by RT-qPCR. PDCD4, AP-1, p-c-Fos, p-c-Jun, and cyclin D1 expressions were analyzed by Western blot. Overexpression of lnc NBAT1 was studied to explore its influence on malignant behaviors of Bel7402 cells and the development of LC in the xenograft mouse model (XMM). The regulation mechanism of lnc NBAT1 in LC was explored by lnc NBAT1 overexpression, miR-21 mimic treatment, or PDCD4 silencing in Bel7402 cells. Lnc NBAT1 expression was downregulated while miR-21 expression was upregulated in LC tissues and cell lines. In comparison with LX-2 cells, the expressions of PDCD4 and AP-1 were downregulated in Bel7402 cells, while those of p-c-Fos, p-c-Jun, and cyclin D1 were upregulated. Further, lnc NBAT1 was found to localize primarily in the cytoplasm of Bel7402 cells. Overexpression of lnc NBAT1 enhanced cell apoptosis, blocked the cell cycle, suppressed malignant behaviors of Bel7402 cells, and inhibited tumor progression in the XMM. Mechanistically, lnc NBAT1 functioned as a competing endogenous RNA (ceRNA) by binding to the downstream target miR-21 to stabilize the expressions of PDCD4 and AP-1, thereby inhibiting malignant behaviors of Bel7402 cells. Lnc NBAT1 suppressed malignant behaviors of LC cells through the miR-21/PDCD4/AP-1 axis. Lnc NBAT1 might be a promising biomarker for LC treatment.
Collapse
Affiliation(s)
- Hongyan Zai
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Wu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yifan Zhou
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Hu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qin Zhu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Li D, Xiong Y, Li M, Long L, Zhang Y, Yan H, Xiang H. STC2 knockdown inhibits cell proliferation and glycolysis in hepatocellular carcinoma through promoting autophagy by PI3K/Akt/mTOR pathway. Arch Biochem Biophys 2024; 761:110149. [PMID: 39271096 DOI: 10.1016/j.abb.2024.110149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/02/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The pathogenesis exploration and timely intervention of hepatocellular carcinoma (HCC) are crucial due to its global impact on human health. As a general tumor biomarker, stanniocalcin 2 (STC2), its role in HCC remains unclear. We aimed to analyze the effect and mechanism of STC2 on HCC. METHODS STC2 expressions in HCC tissues and cell lines were measured. si-STC2 and oe-STC2 transfections were utilized to analyze how STC2 affected cell functions. Functional enrichment analysis of STC2 was performed by Gene Set Enrichment Analysis (GSEA). The regulatory mechanism of STC2 on HCC was investigated using 2-DG, 3-MA, IGF-1, Rap, and LY294002. The impact of STC2 on HCC progression in vivo was evaluated by the tumor formation experiment. RESULTS Higher levels of STC2 expression were observed in HCC tissues and cell lines. Besides, STC2 knockdown reduced proliferation, migration, and invasion, while inducing cell apoptosis. Further analysis indicated a positive correlation between STC2 and glycolysis. STC2 knockdown inhibited glycolysis progression and down-regulated the expressions of PKM2, GLUT1, and HK2 in HCC cells. However, treatment with glycolysis inhibitor (2-DG) prevented oe-STC2 from promoting the growth of HCC cells. Additionally, STC2 knockdown up-regulated the levels of LC3II/LC3I and Beclin1 and reduced the phosphorylation of PI3K, AKT, and mTOR. Treatment with 3-MA, IGF-1, Rap, and LY294002 altered the function of STC2 on proliferation and glycolysis in HCC cells. Tumor formation experiment results revealed that STC2 knockdown inhibited HCC progression. CONCLUSIONS STC2 knockdown inhibited cell proliferation and glycolysis in HCC through the PI3K/Akt/mTOR pathway-mediated autophagy induction.
Collapse
Affiliation(s)
- Ding Li
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Yuanyuan Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Muzi Li
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Lin Long
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Yongjin Zhang
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Huifeng Yan
- The Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Hua Xiang
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China.
| |
Collapse
|
4
|
Lu T, Chen X, Zhang Q, Shang K, Yang X, Xiang W. Vitamin D Relieves Epilepsy Symptoms and Neuroinflammation in Juvenile Mice by Activating the mTOR Signaling Pathway via RAF1: Insights from Network Pharmacology and Molecular Docking Studies. Neurochem Res 2024; 49:2379-2392. [PMID: 38837094 DOI: 10.1007/s11064-024-04176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/15/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Epilepsy is a common neurological disorder, and the exploration of potential therapeutic drugs for its treatment is still ongoing. Vitamin D has emerged as a promising treatment due to its potential neuroprotective effects and anti-epileptic properties. This study aimed to investigate the effects of vitamin D on epilepsy and neuroinflammation in juvenile mice using network pharmacology and molecular docking, with a focus on the mammalian target of rapamycin (mTOR) signaling pathway. Experimental mouse models of epilepsy were established through intraperitoneal injection of pilocarpine, and in vitro injury models of hippocampal neurons were induced by glutamate (Glu) stimulation. The anti-epileptic effects of vitamin D were evaluated both in vivo and in vitro. Network pharmacology and molecular docking analysis were used to identify potential targets and regulatory pathways of vitamin D in epilepsy. The involvement of the mTOR signaling pathway in the regulation of mouse epilepsy by vitamin D was validated using rapamycin (RAPA). The levels of inflammatory cytokines (TNF-α, IL-1β, and IL-6) were assessed by enzyme-linked immunosorbent assay (ELISA). Gene and protein expressions were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. The terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) staining was used to analyze the apoptosis of hippocampal neurons. In in vivo experiments, vitamin D reduced the Racine scores of epileptic mice, prolonged the latency of epilepsy, and inhibited the production of TNF-α, IL-1β, and IL-6 in the hippocampus. Furthermore, network pharmacology analysis identified RAF1 as a potential target of vitamin D in epilepsy, which was further confirmed by molecular docking analysis. Additionally, the mTOR signaling pathway was found to be involved in the regulation of mouse epilepsy by vitamin D. In in vitro experiments, Glu stimulation upregulated the expressions of RAF1 and LC3II/LC3I, inhibited mTOR phosphorylation, and induced neuronal apoptosis. Mechanistically, vitamin D activated the mTOR signaling pathway and alleviated mouse epilepsy via RAF1, while the use of the pathway inhibitor RAPA reversed this effect. Vitamin D alleviated epilepsy symptoms and neuroinflammation in juvenile mice by activating the mTOR signaling pathway via RAF1. These findings provided new insights into the molecular mechanisms underlying the anti-epileptic effects of vitamin D and further supported its use as an adjunctive therapy for existing anti-epileptic drugs.
Collapse
Affiliation(s)
- Tiantian Lu
- School of Pediatrics, Hainan Medical University, Haikou, 571199, China
- Department of Neonatology, Haikou Maternal and Child Health Hospital, Haikou, 570203, China
| | - Xiuling Chen
- Department of Pediatric Medicine, Affiliated Haikou Hospital of Xiangya Medical School Central South University, Haikou, 570208, China
| | - Qin Zhang
- Department of Neurosurgery, Hainan Women and Children's Medical Center, Haikou, 570312, China
| | - Kun Shang
- Institute of Deep-sea Science and Engineering, Chinese Academy of Science, Sanya, 572000, China
| | - Xiaogui Yang
- Department of Neonatology, Haikou Maternal and Child Health Hospital, Haikou, 570203, China
| | - Wei Xiang
- School of Pediatrics, Hainan Medical University, Haikou, 571199, China.
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 570312, China.
- National Health Commission (NHC) Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, 570216, China.
| |
Collapse
|
5
|
Wu J, Lan Z, Li X, He J, Zhang D, Jin T. A novel recombinant adenovirus expressing apoptin and melittin genes kills hepatocellular carcinoma cells and inhibits the growth of ectopic tumor. Invest New Drugs 2024; 42:428-441. [PMID: 38935191 DOI: 10.1007/s10637-024-01453-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
HCC is the most common fatal malignancy. Although surgical resection is the primary treatment strategy, most patients are not eligible for resection due to tumor heterogeneity, underlying liver disease, or comorbidities. Therefore, this study explores the possibility of multi-molecular targeted drug delivery in treating HCC. In this study, we constructed the recombinant adenovirus co-expressing apoptin and melittin (MEL) genes. The inhibitory effect of the recombinant adenovirus on hepatocellular carcinoma cells was detected through experiments on cell apoptosis, migration, invasion, and other factors. The tumor inhibitory effect in vivo was assessed using subcutaneous HCC mice. Results showed that recombinant adenovirus co-expressing anti-tumor genes TAT and apoptin, RGD and MEL can significantly inhibit the proliferation, migration, and invasion of HCC cells by inducing an increase in reactive oxygen species (ROS) levels, upregulation of apoptotic proteins such as Bax, cleaved caspase-3, and cleaved caspase-9, and downregulation of the anti-apoptotic protein Bcl-2. In subcutaneous HCC mice, recombinant adenovirus induced significant apoptosis in tumor, and inhibited tumor growth. In conclusion, recombinant adenovirus co-expressing apoptin and MEL can inhibit the growth and proliferation of tumor cells both in vivo and in vitro.
Collapse
Affiliation(s)
- Jingqiao Wu
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Zhaoyu Lan
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Xin Li
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Jinling He
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
| | - Dongchao Zhang
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China.
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China.
- Tianjin Engineering Technology Center of Livestock Pathogen Detection and Genetic Engineering Vaccine, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China.
| | - Tianming Jin
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin, 300392, China.
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China.
- Tianjin Engineering Technology Center of Livestock Pathogen Detection and Genetic Engineering Vaccine, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China.
| |
Collapse
|
6
|
Wang X, Ye S, Tong L, Gao J, Zhang Y, Qin Y. Inhibition of ROS/caspase-3/GSDME-mediated pyroptosis alleviates high glucose-induced injury in AML-12 cells. Toxicol In Vitro 2024; 98:105840. [PMID: 38723977 DOI: 10.1016/j.tiv.2024.105840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
Diabetic liver injury (DLI) is a chronic complication of the liver caused by diabetes, and its has become one of the main causes of nonalcoholic fatty liver disease (NAFLD). The gasdermin E (GSDME)-dependent pyroptosis signaling pathway is involved in various physiological and pathological processes; however, its role and mechanism in DLI are still unknown. This study was performed to investigate the role of GSDME-mediated pyroptosis in AML-12 cell injury induced by high glucose and to evaluate the therapeutic potential of caspase-3 inhibition for DLI. The results showed that high glucose activated apoptosis by regulating the apoptotic protein levels including Bax, Bcl-2, and enhanced cleavage of caspase-3 and PARP. Notably, some of the hepatocytes treated with high glucose became swollen, accompanied by GSDME-N generation, indicating that pyroptosis was further induced by active caspase-3. Moreover, the effects of high glucose on AML-12 cells could be partly reversed by a reactive oxygen scavenger (NAC) and caspase-3 specific inhibitor (Z-DEVD-FMK), which suggests high glucose induced GSDME-dependent pyroptosis in AML-12 cells through increasing ROS levels and activating caspase-3. In conclusion, our results show that high glucose can induce pyroptosis in AML-12 cells, at least in part, through the ROS/caspase-3/GSDME pathway,and inhibition of caspase-3 can ameliorate high glucose-induced hepatocyte injury, providing an important basis for clarifying the pathogenesis and treatment of DLI.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Shengying Ye
- Department of Physiology and Pathophysiology, School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Linge Tong
- Department of Physiology and Pathophysiology, School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Jingwen Gao
- Department of Physiology and Pathophysiology, School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Yixin Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Yan Qin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Dali University, Dali, Yunnan 671000, China.
| |
Collapse
|
7
|
Li YC, Wei ZY, Chai B, Pan Z, Zhang SZ, Li H, Wang JL, Ye XJ. Highly adhesive self-reinforce hydrogel for the amelioration of intervertebral disc degeneration: Eliminating reactive oxygen species and regulating extracellular matrix. COMPOSITES PART B: ENGINEERING 2024; 275:111280. [DOI: 10.1016/j.compositesb.2024.111280] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
8
|
Wu Y, Li M, Ying H, Gu Y, Zhu Y, Gu Y, Huang L. Mitochondrial quality control alterations and placenta-related disorders. Front Physiol 2024; 15:1344951. [PMID: 38390447 PMCID: PMC10883312 DOI: 10.3389/fphys.2024.1344951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/17/2024] [Indexed: 02/24/2024] Open
Abstract
Mitochondria are ubiquitous in eukaryotic cells. Normal maintenance of function is the premise and basis for various physiological activities. Mitochondrial dysfunction is commonly observed in a wide range of pathological conditions, such as neurodegenerative, metabolic, cardiovascular, and various diseases related to foetal growth and development. The placenta is a highly energy-dependent organ that acts as an intermediary between the mother and foetus and functions to maintain foetal growth and development. Recent studies have demonstrated that mitochondrial dysfunction is associated with placental disorders. Defects in mitochondrial quality control mechanisms may lead to preeclampsia and foetal growth restriction. In this review, we address the quality control mechanisms of mitochondria and the relevant pathologies of mitochondrial dysfunction in placenta-related diseases, such as preeclampsia and foetal growth restriction. This review also investigates the relation between mitochondrial dysfunction and placental disorders.
Collapse
Affiliation(s)
- Yamei Wu
- Wuxi Maternity and Child Healthcare Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Medical College of Nanjing Medical University, Wuxi, China
| | - Meng Li
- Wuxi Maternity and Child Healthcare Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Medical College of Nanjing Medical University, Wuxi, China
| | - Hao Ying
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Gu
- Wuxi Maternity and Child Healthcare Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Medical College of Nanjing Medical University, Wuxi, China
| | - Yunlong Zhu
- Wuxi Maternity and Child Healthcare Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Medical College of Nanjing Medical University, Wuxi, China
| | - Yanfang Gu
- Wuxi Maternity and Child Healthcare Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Medical College of Nanjing Medical University, Wuxi, China
| | - Lu Huang
- Wuxi Maternity and Child Healthcare Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
- Wuxi Clinical Medical College of Nanjing Medical University, Wuxi, China
| |
Collapse
|
9
|
Guo C, Liu Y, Zhao Z, Wu Y, Kong Q, Wang Y. Regulating inflammation and apoptosis: A smart microgel gene delivery system for repairing degenerative nucleus pulposus. J Control Release 2024; 365:1004-1018. [PMID: 38128882 DOI: 10.1016/j.jconrel.2023.12.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
The progression of intervertebral disc degeneration (IDD) is attributed to the gradual exacerbation of cellular apoptosis and impaired extracellular matrix (ECM) synthesis, both of which are induced by progressive inflammation. Therefore, it is crucial to address the inflammatory microenvironment and rectify the excessive apoptosis of nucleus pulposus cells (NPCs) to achieve intervertebral disc (IVD) regeneration. In this study, we devised a smart microgel gene delivery system that incorporates functionalized gene nanoparticles (NPs) for the purpose of IVD regeneration. siGrem1 was loaded into the NPs to enhance their antiapoptotic ability and protective effects. Furthermore, the encapsulation of HADA further endows the NPs (referred to as HSGN) with targeted delivery and anti-inflammatory effects, as well as reactive oxygen species (ROS) scavenging capacities. To create an microenvironment-responsive microgel system, phenylboronic acid-functionalized microspheres (referred to as M.S.) were fabricated and dynamically loaded with the HSGN. This microgel system (MHSGN), which is highly biocompatible, enables the sustained release of siGrem1, effectively modulating inflammation, scavenging ROS, and alleviating apoptosis in NPCs. These multifunctional capabilities promote the restoration of metabolic homeostasis within the nucleus pulposus ECM, ultimately leading to delayed IDD.
Collapse
Affiliation(s)
- Chuan Guo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Yuheng Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Zhen Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Ye Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041
| | - Qingquan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041.
| | - Yu Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610041.
| |
Collapse
|
10
|
Wang KD, Zhu ML, Qin CJ, Dong RF, Xiao CM, Lin Q, Wei RY, He XY, Zang X, Kong LY, Xia YZ. Sanguinarine induces apoptosis in osteosarcoma by attenuating the binding of STAT3 to the single-stranded DNA-binding protein 1 (SSBP1) promoter region. Br J Pharmacol 2023; 180:3175-3193. [PMID: 37501645 DOI: 10.1111/bph.16202] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/19/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Osteosarcoma, a primary malignant bone tumour prevalent among adolescents and young adults, remains a considerable challenge despite protracted progress made in enhancing patient survival rates over the last 40 years. Consequently, the development of novel therapeutic approaches for osteosarcoma is imperative. Sanguinarine (SNG), a compound with demonstrated potent anticancer properties against various malignancies, presents a promising avenue for exploration. Nevertheless, the intricate molecular mechanisms underpinning SNG's actions in osteosarcoma remain elusive, necessitating further elucidation. EXPERIMENTAL APPROACH Single-stranded DNA-binding protein 1 (SSBP1) was screened out by differential proteomic analysis. Apoptosis, cell cycle, reactive oxygen species (ROS) and mitochondrial changes were assessed via flow cytometry. Western blotting and quantitative real-time reverse transcription PCR (qRT-PCR) were used to determine protein and gene levels. The antitumour mechanism of SNG was explored at a molecular level using chromatin immunoprecipitation (ChIP) and dual luciferase reporter plasmids. KEY RESULTS Our investigation revealed that SNG exerted an up-regulated effect on SSBP1, disrupting mitochondrial function and inducing apoptosis. In-depth analysis uncovered a mechanism whereby SNG hindered the JAK/signal transducer and activator of transcription 3 (STAT3) signalling pathway, relieved the inhibitory effect of STAT3 on SSBP1 transcription, and inhibited the downstream PI3K/Akt/mTOR signalling axis, ultimately activating apoptosis. CONCLUSIONS AND IMPLICATIONS The study delved further into elucidating the anticancer mechanism of SNG in osteosarcoma. Notably, we unravelled the previously undisclosed apoptotic potential of SSBP1 in osteosarcoma cells. This finding holds substantial promise in advancing the development of novel anticancer drugs and identification of therapeutic targets.
Collapse
Affiliation(s)
- Kai-Di Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Miao-Lin Zhu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Cheng-Jiao Qin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rui-Fang Dong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Cheng-Mei Xiao
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qing Lin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rong-Yuan Wei
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiao-Yu He
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin Zang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuan-Zheng Xia
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
Zhu Y, Wang S, Niu P, Chen H, Zhou J, Jiang L, Li D, Shi D. Raptor couples mTORC1 and ERK1/2 inhibition by cardamonin with oxidative stress induction in ovarian cancer cells. PeerJ 2023; 11:e15498. [PMID: 37304865 PMCID: PMC10257395 DOI: 10.7717/peerj.15498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Background A balance on nutrient supply and redox homeostasis is required for cell survival, and increased antioxidant capacity of cancer cells may lead to chemotherapy failure. Objective To investigate the mechanism of anti-proliferation of cardamonin by inducing oxidative stress in ovarian cancer cells. Methods After 24 h of drug treatment, CCK8 kit and wound healing test were used to detect cell viability and migration ability, respectively, and the ROS levels were detected by flow cytometry. The differential protein expression after cardamonin administration was analyzed by proteomics, and the protein level was detected by Western blotting. Results Cardamonin inhibited the cell growth, which was related to ROS accumulation. Proteomic analysis suggested that MAPK pathway might be involved in cardamonin-induced oxidative stress. Western blotting showed that cardamonin decreased Raptor expression and the activity of mTORC1 and ERK1/2. Same results were observed in Raptor KO cells. Notably, in Raptor KO cells, the effect of cardamonin was weakened. Conclusion Raptor mediated the function of cardamonin on cellular redox homeostasis and cell proliferation through mTORC1 and ERK1/2 pathways.
Collapse
|
12
|
Xu X, Liu Y, Gao J, Shi X, Yan Y, Yang N, Wang Q, Zhang Q. GRAMD4 regulates PEDV-induced cell apoptosis inhibiting virus replication via the endoplasmic reticulum stress pathway. Vet Microbiol 2023; 279:109666. [PMID: 36738512 DOI: 10.1016/j.vetmic.2023.109666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/14/2023] [Accepted: 01/22/2023] [Indexed: 01/25/2023]
Abstract
Porcine epidemic diarrhea (PED) caused by the porcine epidemic diarrhea virus (PEDV) has caused huge losses in the swine industry worldwide. Glucosyltransferase Rab-like GTPase activator and myotubularin domain containing 4 (GRAMD4) is a proapoptotic protein, which replaced p53 inducing mitochondrial apoptosis. However, the relationship between GRAMD4 and PEDV has not been reported. Here, we aimed to investigate the potential role of GRAMD4 during PEDV infection. In this study, we used co-immunoprecipitation (co-IP) and mass spectrometry to identify GRAMD4 interaction with PEDV non-structural protein 6 (NSP6). Immunoprecipitation and laser confocal microscopy were utilized to demonstrate that GRAMD4 interacts with NSP6. NSP6 reduces GRAMD4 production through PERK and IRE1 pathway-mediated apoptosis. We demonstrated that overexpression of GRAMD4 effectively impaired the replication of PEDV, whereas knockdown of GRAMD4 facilitated the replication of PEDV. Overexpression of GRAMD4 increased GRP78, phosphorylated PERK (p-PERK), phosphorylated IRE1(p-IRE1) levels, promoted CHOP, phosphorylated JNK (p-JNK), Bax expression, caspase 9 and caspase 3 cleavage, and inhibited Bcl-2 production. Knockdown of GRAMD4 has the opposite effect. Finally, deletion of the GRAM domain of GRAMD4 cannot cause endoplasmic reticulum stress (ER stress)-mediated apoptosis and inhibit virus replication. In conclusion, these studies revealed the mechanism by which GRAMD4 was associated with ER stress and apoptosis regulating PEDV replication. NSP6 acted as a potential down-regulator of GRAMD4 and promoted the degradation of GRAMD4. GRAMD4 played a role in facilitating apoptosis and restricting virus replication, and the GRAM domain was required. These findings provided a reference for host-PEDV interactions and offered the possibility for PEDV decontamination and prevention.
Collapse
Affiliation(s)
- Xingang Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yi Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jie Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaojie Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yuchao Yan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Naling Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Quanqiong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qi Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
13
|
Mitophagy Effects of Protodioscin on Human Osteosarcoma Cells by Inhibition of p38MAPK Targeting NIX/LC3 Axis. Cells 2023; 12:cells12030395. [PMID: 36766737 PMCID: PMC9913878 DOI: 10.3390/cells12030395] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Protodioscin (PD) is a steroidal saponin with various pharmacological activities, including neuro-protective, anti-inflammatory, and anti-tumor activities. However, the effect of PD on human osteosarcoma (OS) cells is unclear. In this study, we found that PD significantly inhibits the growth of human HOS and 143B OS cells through the upregulation of apoptotic-related proteins (cleaved caspase-3, cleaved caspase-9, and cleaved PARP) and mitophagy-related proteins (LC3B and NIX), which contribute to the induction of apoptosis, and MMP (mitochondrial membrane potential) dysfunction and mitophagy. The inhibition of LC3 or NIX was shown to decrease apoptosis and mitophagy in PD-treated OS cells. The knockdown of p38MAPK by siRNA decreased mitochondrial dysfunction, autophagy, mitophagy, and the NIX/LC3B expression in the PD-treated OS cells. A binding affinity analysis revealed that the smaller the KD value (-7.6 Kcal/mol and -8.9 Kcal/mol, respectively), the greater the binding affinity in the PD-NIX and PD-LC3 complexes. These findings show the inhibitory effects of PD-induced mitophagy in human OS cells and may represent a novel therapeutic strategy for human OS, by targeting the NIX/LC3 pathways.
Collapse
|
14
|
Liu C, Sun S, Mao J. Water-soluble Yb 3+, Er 3+ codoped NaYF 4 nanoparticles induced SGC-7901 cell apoptosis through mitochondrial dysfunction and ROS-mediated ER stress. Hum Exp Toxicol 2023; 42:9603271231188493. [PMID: 37419518 DOI: 10.1177/09603271231188493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
BACKGROUND Nanoparticles are potential luminescent probes; among them, upconversion nanoparticles (UCNP) are currently being developed as fluorescent probes for biomedical applications. However, the molecular mechanisms of UCNP in human gastric cell lines remain poorly understood. Here, we aimed to examine UCNP cytotoxicity to SGC-7901 cells and explore its underlying mechanisms. METHODS The effects of 50-400 μg/mL UCNP on human gastric adenocarcinoma (SGC-7901) cells were investigated. Flow cytometry was used to evaluate reactive oxygen species (ROS), mitochondrial membrane potential (ΔΨm), intracellular Ca2+ levels, and apoptosis. Activated caspase-3 and nine activities were measured; meanwhile, cytochrome C (Cyt C) in the cytosol and B-cell lymphoma 2 (Bcl-2), Bcl-2 associated X protein (Bax), protein kinase B (Akt), phosphorylated-Akt (p-Akt), 78 kDa glucose-regulated protein (GRP78), 94 kDa glucose-regulated protein (GRP94), calpain-1, and calpain-2 protein levels were also detected. RESULTS UCNP inhibited the viability of SGC-7901 cells in a concentration- and time-dependent manner and increased the proportion of cell apoptosis. Exposure to UCNP enhanced the ratio of Bax/Bcl-2, elevated the level of ROS, decreased ΔΨm, increased intracellular Ca2+ and Cyt C protein levels, decreased the levels of phosphorylated Akt, increased the activity of caspase-3 and caspase-9, and upregulated the protein expression of GRP-78, GRP-94, calpain-1 and calpain-2 in SGC-7901 cells. CONCLUSION UCNP induced SGC-7901 cell apoptosis by promoting mitochondrial dysfunction and ROS-mediated endoplasmic reticulum (ER) stress, initiating the caspase-9/caspase-3 cascade.
Collapse
Affiliation(s)
- Chen Liu
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Shaoqiang Sun
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Jingwei Mao
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| |
Collapse
|
15
|
Yang K, Feng S, Luo Z. Oncolytic Adenovirus, a New Treatment Strategy for Prostate Cancer. Biomedicines 2022; 10:biomedicines10123262. [PMID: 36552019 PMCID: PMC9775875 DOI: 10.3390/biomedicines10123262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Prostate cancer is the most common cancer and one of the leading causes of cancer mortality in males. Androgen-deprivation therapy (ADT) is an effective strategy to inhibit tumour growth at early stages. However, 10~50% of cases are estimated to progress to metastatic castration-resistant prostate cancer (mCRPC) which currently lacks effective treatments. Clinically, salvage treatment measures, such as endocrine therapy and chemotherapy, are mostly used for advanced prostate cancer, but their clinical outcomes are not ideal. When the existing clinical therapeutic methods can no longer inhibit the development of advanced prostate cancer, human adenovirus (HAdV)-based gene therapy and viral therapy present promising effects. Pre-clinical studies have shown its powerful oncolytic effect, and clinical studies are ongoing to further verify its effect and safety in prostate cancer treatment. Targeting the prostate by HAdV alone or in combination with radiotherapy and chemotherapy sheds light on patients with castration-resistant and advanced prostate cancer. This review summarizes the advantages of oncolytic virus-mediated cancer therapy, strategies of HAdV modification, and existing preclinical and clinical investigations of HAdV-mediated gene therapy to further evaluate the potential of oncolytic adenovirus in prostate cancer treatment.
Collapse
Affiliation(s)
- Kaiyi Yang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
- Correspondence: (K.Y.); (Z.L.)
| | - Shenghui Feng
- Provincial Key Laboratory of Tumour Pathogens and Molecular Pathology, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhijun Luo
- Provincial Key Laboratory of Tumour Pathogens and Molecular Pathology, Queen Mary School, Nanchang University, Nanchang 330031, China
- Correspondence: (K.Y.); (Z.L.)
| |
Collapse
|