1
|
Zhang Z, Yu W, He X, Ying J, Liu Y, Chen M, Fu D, Shen A, Liang X. In-depth characterization of quinolizidine alkaloids via highly efficient enrichment and multi-stage mass spectrometry. Talanta 2025; 293:128141. [PMID: 40249983 DOI: 10.1016/j.talanta.2025.128141] [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: 01/23/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 04/20/2025]
Abstract
The identification of new natural products is a critical step in the discovery of lead compounds, and is a prerequisite for the development of novel pharmaceuticals. Traditional Chinese medicines (TCMs) are renowned for their diverse pharmacological activities, and represent a valuable source of novel chemicals. The quinolizidine alkaloids are abundant in plants of the Fabaceae family and exhibit significant bioactivities, including anticancer and anti-inflammatory effects. However, the structural diversity of quinolizidine alkaloids and the chemical complexity associated with TCMs pose great challenges in the discovery of novel quinolizidine alkaloids. In the present study, Sophora flavescens Ait. was used as a model to establish a comprehensive strategy for characterizing quinolizidine alkaloids. A simple and selective method was developed using an FC8HL column for the efficient enrichment of polar quinolizidine alkaloids. Through the integration of high-resolution multi-stage mass spectrometry and feature-based molecular networking, the enriched alkaloids were thoroughly characterized, including matrine-type, cystine-type, aloperine-type, anagyrine-type, lupinine-type, and dimeric species. A total of 186 quinolizidine alkaloids were identified, including 131 newly discovered compounds. A series of novel substituents was also identified for the first time. The findings of this study not only deepen our understanding of the structural diversity of quinolizidine alkaloids, but they also offer a novel research strategy for the comprehensive characterization of quinolizidine alkaloids in other plants, potentially facilitating the discovery of new drug candidates.
Collapse
Affiliation(s)
- Zhen Zhang
- School of Biological Engineering, Dalian Polytechnic University, Dalian, 116034, China; State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Wenyi Yu
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Xiao He
- Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Jinqin Ying
- Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yanfang Liu
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Ming Chen
- School of Biological Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Dongmei Fu
- School of Biological Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Aijin Shen
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China.
| | - Xinmiao Liang
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China; Jiangxi Provincial Key Laboratory for Pharmacodynamic Material Basis of Traditional Chinese Medicine, Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| |
Collapse
|
2
|
Wang Z, Li X, Zhao L, Liu S, Du J, Jia X, Ge L, Xu J, Cui K, Ga Y, Wang J, Xia X. Matrine Restores Colistin Efficacy Against mcr-1-Carrying Escherichia coli. Molecules 2025; 30:2122. [PMID: 40430295 PMCID: PMC12113751 DOI: 10.3390/molecules30102122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The emergence of mcr-1-mediated colistin resistance has become a critical global health concern, highlighting the urgent need for innovative approaches to restore colistin's therapeutic potential. In this study, we evaluated the antibacterial activity of four matrine-type alkaloids-namely, matrine, oxymatrine, sophocarpine, and sophoramine-against mcr-1-positive Escherichia coli. While these alkaloids showed limited efficacy when used alone, the combination of matrine with colistin exhibited remarkable synergistic effects, as demonstrated by checkerboard assays and time-kill curve analyses. The matrine-colistin combination caused minimal erythrocyte damage while effectively attenuating resistance development in vitro. This synergy was further corroborated in a murine infection model, where the combination significantly reduced bacterial loads in target tissues. Mechanistic studies revealed that the matrine-colistin combination enhances antimicrobial activity by disrupting bacterial membrane integrity, increasing intracellular colistin accumulation, and triggering reactive oxygen species-mediated oxidative damage. Collectively, these findings highlight the potential of matrine as a promising adjuvant to overcome colistin resistance, providing a novel therapeutic approach to address the challenge of infections cause by multidrug-resistant Gram-negative bacteria.
Collapse
Affiliation(s)
- Zhinan Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Xiaowei Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Liang Zhao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Saiwa Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Jingjing Du
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Xi Jia
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Lirui Ge
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Jian Xu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Kexin Cui
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Yu Ga
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| | - Jinxiu Wang
- Hainan Provincial Animal Disease Prevention and Control Center, Haikou 571100, China
| | - Xi Xia
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Z.W.); (X.L.); (L.Z.); (S.L.); (J.D.); (X.J.); (L.G.); (J.X.); (K.C.); (Y.G.)
| |
Collapse
|
3
|
Liu X, Li Y, Yuan C, Zhao Y, Zhou L, Yan Y, Ren J, Liu Q. Sophocarpine suppresses MAPK-mediated inflammation by restoring gut microbiota in colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156833. [PMID: 40393246 DOI: 10.1016/j.phymed.2025.156833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/18/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Colorectal cancer (CRC), as one of the most common cancers globally, poses a significant challenge to public health due to its high incidence and mortality rates. This underscores the need for continuous exploration of new therapeutic targets and effective drugs. Sophocarpine (SC), a natural compound derived from traditional Chinese medicine, holds considerable therapeutic potential in the treatment of CRC, however, the relevant mechanisms remains unclear. PURPOSE This study aims to explore the anti-tumor effects of SC against CRC by modulating gut microbiota, and uncover potential mechanisms linking SC's therapeutic effects to gut microbiota regulation by analyzing the impact of SC on microbiota composition and CRC progression. MATERIAL This study explores the impact of SC on the gut microbiota in CRC by constructing subcutaneous xenograft tumors of CRC and integrating 16S rRNA sequencing and RNA transcriptomic sequencing. The fecal microbiota transplantation (FMT) mouse model was used to validate the biological function of SC in correcting gut microbiota dysbiosis to treat CRC. Subsequently, we conducted in vitro studies on the molecular mechanisms by which SC regulates the gut microbiota as an effective hallmark of CRC treatment, using lipopolysaccharide (LPS) to simulate an inflammatory gut microbiota environment and P38 MAPK knockdown cell line. RESULTS SC significantly inhibited CRC cell proliferation with IC50 values of 2.547±0.256 μM for HCT116 and 2.851±0.332 μM for LoVo cells. In vivo experiments demonstrated that SC effectively suppressed tumor growth in xenograft models. 16S rRNA sequencing revealed that SC modulated gut microbiota composition, particularly affecting Bacteroides and Alistipes populations. SC significantly reduced the levels of inflammatory factors and inhibited the MAPK signaling pathway, as evidenced by decreased p-JNK, p-p38 MAPK, and p-NF-κB p65 expression. CONCLUSIONS Current clinical practice still lacks effective therapeutic agents targeting CRC through gut microbiota modulation. This study presents the first evidence that SC, a natural compound, exhibits dual-action therapeutic efficacy against CRC progression by simultaneously modulating gut microbial composition and suppressing MAPK pathway-mediated inflammatory responses. These findings highlight SC's novel therapeutic potential as a promising microbiota-regulating candidate for CRC intervention, offering an innovative approach that bridges microbial ecology with cancer signaling pathways.
Collapse
Affiliation(s)
- Xiangjun Liu
- Laboratory Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Yu Li
- Laboratory Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Chenyue Yuan
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Yong Zhao
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Lin Zhou
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Yuting Yan
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Jianlin Ren
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China.
| | - Qingzhong Liu
- Laboratory Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China.
| |
Collapse
|
4
|
Su K, Tang M, Wu J, Ye N, Jiang X, Zhao M, Zhang R, Cai X, Zhang X, Li N, Peng J, Lin L, Wu W, Ye H. Mechanisms and therapeutic strategies for NLRP3 degradation via post-translational modifications in ubiquitin-proteasome and autophagy lysosomal pathway. Eur J Med Chem 2025; 289:117476. [PMID: 40056798 DOI: 10.1016/j.ejmech.2025.117476] [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: 12/13/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
The NLRP3 inflammasome is crucial for immune responses. However, its overactivation can lead to severe inflammatory diseases, underscoring its importance as a target for therapeutic intervention. Although numerous inhibitors targeting NLRP3 exist, regulating its degradation offers an alternative and promising strategy to suppress its activation. The degradation of NLRP3 is primarily mediated by the proteasomal and autophagic pathways. The review not only elaborates on the traditional concepts of ubiquitination and NLRP3 degradation but also investigates the important roles of indirect regulatory modifications, such as phosphorylation, acetylation, ubiquitin-like modifications, and palmitoylation-key post-translational modifications (PTMs) that influence NLRP3 degradation. Additionally, we also discuss the potential targets that may affect NLRP3 degradation during the proteasomal and autophagic pathways. By unraveling these complex regulatory mechanisms, the review aims to enhance the understanding of NLRP3 regulation and its implications for developing therapeutic strategies to combat inflammatory diseases.
Collapse
Affiliation(s)
- Kaiyue Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wu
- Key Laboratory of Hydrodynamics (Ministry of Education), School of Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Neng Ye
- Scaled Manufacturing Center of Biological Products, Management Office of National Facility for Translational Medicine, West China Hospital, Sichuan University Chengdu 610041, China
| | - Xueqin Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Laboratory of Metabolomics and Drug-induced Liver Injury, Department of Gastroenterology & Hepatology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruijia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinlu Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Peng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haoyu Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
5
|
Wei S, Xiao J, Ju F, Hu Z. Aloperine protects the testis against testicular ischemia/reperfusion injury in rats. Andrology 2025; 13:934-954. [PMID: 39253755 DOI: 10.1111/andr.13750] [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: 01/19/2024] [Revised: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Testicular torsion/detorsion can cause testis loss and infertility. Aloperine is a major active alkaloid extracted from Sophora alopecuroides Linn. It has been shown to have organ-protective effects. However, the effects of aloperine on the testis and its underlying mechanisms remain unclear. OBJECTIVES This study investigated the effect of aloperine on testicular torsion/detorsion injury in rats. MATERIALS AND METHODS Male Sprague-Dawley rats were randomized to the sham-operated (sham), testicular I/R (TI/R), or aloperine preconditioning (ALOPre) or postconditioning (ALOPost) groups. All rats except for the sham-operated rats were subjected to 3 h of right spermatic cord torsion (720°, clockwise), followed by 3 h of detorsion. Aloperine (10 mg/kg) was intravenously administered before testicular torsion (ALOPre) or at the onset of testicular detorsion (ALOPost). The therapeutic efficacy of aloperine was evaluated by histological analysis, oxidative stress evaluation, inflammatory response examination, apoptosis analysis, protein analysis, and immunohistological assessment. RESULTS Compared with TI/R, aloperine protected both the ipsilateral and contralateral testes against unilateral testicular I/R, as evidenced by a reduced testicular weight to body weight (TW/BW) ratio (ALOPre: p = 0.0037; ALOPost: p = 0.0021) and volume (ALOPre: p = 0.0020; ALOPost: p = 0.0009), less structural damage with better Johnsen (ALOPre: p = 0.0013; ALOPost: p = 0.0021), and Cosentino scores (ALOPre: p < 0.0001; ALOPost: p < 0.0001), increased mean seminiferous tubule diameter and mean seminiferous tubule epithelial height, decreased testicular apoptosis, and less oxidative stress and inflammatory response. In addition, aloperine significantly stimulated the phosphorylation of signal transducer and activator of transcription (STAT)-3 in the ipsilateral testes following detorsion. Administration of Ag490 suppressed STAT-3 phosphorylation, thereby abrogating the protective effects exerted by aloperine on the ipsilateral testis. DISCUSSION AND CONCLUSION Aloperine has a strong testicular protective effect on the ipsilateral and contralateral testes after testicular torsion/detorsion. This aloperine-induced ipsilateral testicular protection is mediated via the STAT-3 signaling pathway.
Collapse
Affiliation(s)
- Shichao Wei
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junshen Xiao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Ju
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaoyang Hu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Si F, Wang Q, Chen F, Lu X. Sophocarpine inhibits the proliferation and induces apoptosis of glioblastoma cells through regulating the miR-21/PTEN/PI3K/AKT axis. Discov Oncol 2025; 16:138. [PMID: 39921785 PMCID: PMC11807034 DOI: 10.1007/s12672-025-01839-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/20/2025] [Indexed: 02/10/2025] Open
Abstract
Sophocarpine (SC) has been reported to suppress tumorigenesis. But the effect of SC on glioblastoma (GBM) is unknown. This study explored the anti-proliferation and pro-apoptosis effects of SC on GBM cells and the molecular mechanism. Different concentrations of SC were used to treat human astrocyte NHA and GBM cells lines LN229 and SF539. CCK-8 was applied to analyze cell toxicity and proliferation. qRT-PCR and western blot were used to measure RNA and protein expressions, respectively. Cell cycle and cell apoptosis were determined by flow cytometry assay. The results indicated that SC inhibited proliferation and induced apoptosis of LN229 and SF539 cells in a dose-dependent manner. The arrest of the G0/G1 phase of GBM cells was increased after SC treatment. Moreover, SC downregulated miR-21 expression and upregulated PTEN expression in GBM cells. Overexpression of miR-21 partly abrogated the anti-proliferation and pro-apoptosis effects of SC on GBM cells, while exogenous PTEN partially eliminated the pro-proliferation and anti-apoptosis effects of miR-21 on GBM cells. Furthermore, SC treatment decreased the levels of PI3K/AKT pathway-related p-PI3K, p-AKT and PIP3 in GBM cells. The PI3K/AKT pathway activator 740Y-P partially reversed the reduced cell proliferation and enhanced cell apoptosis in SC-treated GBM cells. Significantly, we verified that SC suppressed the proliferation and enhanced apoptosis of GBM cells via inhibiting miR-21 while it was not entirely dependent on upregulation PTEN. Consequently, the potential mechanism of SC in induction apoptosis of GBM cells was verified, which might provide a new method for GBM treatment.
Collapse
Affiliation(s)
- Feng Si
- Department of Neurosurgery, People's Hospital Affiliated to Shandong First Medical University, Jinan, 271100, Shandong, China
| | - Qian Wang
- Department of Orthopedics, People's Hospital Affiliated to Shandong First Medical University, Jinan, 271100, Shandong, China
| | - Fei Chen
- Department of Neurosurgery, People's Hospital Affiliated to Shandong First Medical University, Jinan, 271100, Shandong, China
| | - Xiangdong Lu
- Department of Neurosurgery, People's Hospital Affiliated to Shandong First Medical University, Jinan, 271100, Shandong, China.
- Nanchang University, Nanchang, 330031, Jiangxi, China.
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
7
|
Wei S, Xiao J, Ju F, Li J, Liu T, Hu Z. Aloperine Attenuates Hepatic Ischemia/Reperfusion-Induced Liver Injury via STAT-3 Signaling in a Murine Model. J Pharmacol Exp Ther 2024; 391:51-63. [PMID: 39164092 DOI: 10.1124/jpet.123.001992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024] Open
Abstract
Hepatic ischemia/reperfusion (I/R) damage is one of the most common side effects of liver surgery. This pathophysiological process may lead to excessive hepatic damage. Aloperine is an active ingredient isolated from Sophora alopecuroides Linn and has a variety of therapeutic effects, including organ protection. However, the hepatoprotective effect of aloperine against hepatic I/R damage has not yet been determined. C57BL/6 mice were allocated to the sham-operated (sham), hepatic ischemia/reperfusion (I/R), and aloperine groups. The mice were exposed to 30 min of hepatic hilum occlusion. Then a 3-h reperfusion was performed. Mice in the sham group underwent sham surgery. Hepatic injury was evaluated by plasma aspartate aminotransferase (AST) and transaminase alanine aminotransferase (ALT) levels, histological evaluation, cell apoptosis, the number of activated inflammatory cells, and the expression levels of inflammatory cytokines, including tumor necrosis factor-α and interleukin-6. The protein phosphorylation status of the reperfusion-associated survival pathways was evaluated. Mice with hepatic I/R injury presented increased plasma ALT and AST levels, increased hepatic apoptosis, abnormal histological structure, and elevated inflammatory responses. However, aloperine ameliorated hepatic I/R-induced injury. Moreover, aloperine enhanced the level of signal transducer and activator of transcription (STAT)-3 phosphorylation after I/R. Ag490, an agent that inhibits STAT-3 activity, abolished aloperine-induced STAT-3 phosphorylation and liver protection. Aloperine ameliorates hepatic I/R-induced liver injury via a STAT-3-mediated protective mechanism. Patients with hepatic I/R injury may benefit from aloperine treatment. SIGNIFICANCE STATEMENT: Hepatic I/R can cause excessive liver damage. This study revealed that aloperine, an active component isolated from Sophora alopecuroides Linn, ameliorates hepatic I/R injury and related liver damage in vivo. The underlying protective mechanism may involve the STAT-3 signaling pathway. These findings may lead to the development of a novel approach for treating hepatic I/R damage in clinical practice.
Collapse
Affiliation(s)
- Shichao Wei
- Department of Anesthesiology (S.W., J.X., F.J., J.L.) and Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology (S.W., J.X., F.J., J.L., T.L., Z.H.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junshen Xiao
- Department of Anesthesiology (S.W., J.X., F.J., J.L.) and Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology (S.W., J.X., F.J., J.L., T.L., Z.H.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Ju
- Department of Anesthesiology (S.W., J.X., F.J., J.L.) and Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology (S.W., J.X., F.J., J.L., T.L., Z.H.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaxue Li
- Department of Anesthesiology (S.W., J.X., F.J., J.L.) and Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology (S.W., J.X., F.J., J.L., T.L., Z.H.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Liu
- Department of Anesthesiology (S.W., J.X., F.J., J.L.) and Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology (S.W., J.X., F.J., J.L., T.L., Z.H.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaoyang Hu
- Department of Anesthesiology (S.W., J.X., F.J., J.L.) and Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology (S.W., J.X., F.J., J.L., T.L., Z.H.), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|