1
|
Zhu J, Wen N, Chen W, Yu H. Mitochondrial ribosomal proteins: potential targets for cancer prognosis and therapy. Front Oncol 2025; 15:1586137. [PMID: 40371222 PMCID: PMC12074914 DOI: 10.3389/fonc.2025.1586137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025] Open
Abstract
Mitochondrial ribosomal proteins (MRPs) are essential components of mitochondrial ribosomes, responsible for translating proteins encoded by mitochondrial DNA and maintaining mitochondrial energy metabolism and function. Emerging evidence suggests that MRPs exhibit significant expression changes in multiple cancer types, profoundly affecting tumor biology through modulating oxidative stress levels, inducing metabolic reprogramming, disrupting cell cycle regulation, inhibiting apoptosis, promoting mitophagy, and remodeling the tumor microenvironment. Specifically, MRPs have been implicated in tumor cell proliferation, migration, invasion, and apoptosis, highlighting their potential as therapeutic targets. This review summarizes the multifaceted roles of MRPs in cancer, focusing on their impact on the tumor microenvironment and their potential as prognostic biomarkers and therapeutic targets. We also explore the implications of MRPs in precision oncology, particularly in patient stratification and the design of metabolic targeted therapies, offering new insights and research directions for the precise prevention and treatment of cancer.
Collapse
Affiliation(s)
- Jianqing Zhu
- Postgraduate Department, Hebei North University, Zhangjiakou, China
| | - Na Wen
- Department of Obstetrics and Gynecology, The Eighth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Wen Chen
- Department of Pathology, The Eighth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Haotian Yu
- Department of Obstetrics and Gynecology, The Eighth Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
2
|
Liu W, Lu D, Jia S, Yang Y, Meng F, Du Y, Yang Y, Yuan L, Nan Y. Molecular mechanism of Gancao Xiexin Decoction regulating EMT and suppressing hepatic metastasis of gastric cancer via the TGF-β1/SMAD pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119430. [PMID: 39900270 DOI: 10.1016/j.jep.2025.119430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gastric cancer (GC) is a highly malignant tumor of the digestive tract, posing a significant menace to human health. Gancao Xiexin Decoction (GCXXD), being a traditional Chinese medicine (TCM), has a good effect on inhibiting the proliferation and metastasis of GC. However, its mechanisms still need further investigation. AIM OF STUDY To investigate the mechanism by which GCXXD inhibits GC metastasis through network pharmacology, and to verify through in vivo and in vitro experiments. MATERIALS AND METHODS The TCMSP and GEO databases, in combination with UPLC-MS/MS techniques, were employed to identify the hub genes, active ingredients, and critical pathways of GCXXD in the treatment of GC. Subsequently, molecular docking was conducted on both the hub genes and the core components. Finally, based on the results of the bioinformatics analysis, the role of GCXXD in inhibiting liver metastasis of GC was elucidated through in vivo and in vitro experiments, including scratch assays, Transwell assays, HE staining, immunohistochemistry, in vivo live imaging, qRT-PCR, and Western blotting. RESULTS Utilizing UPLC-MS/MS and network pharmacology, we identified 20 active ingredients and 5 hub targets in the treatment of GC by GCXXD. Through KEGG analyses, GCXXD treatment of GC could through the TGF-beta pathway. In vivo and in vitro experiments, GCXXD downregulated the mRNA and protein expression level of hub genes involved in the TGF-β1/SMAD pathway and the EMT process. Additionally, GCXXD significantly reduced the incidence of liver metastases in GC. CONCLUSION GCXXD inhibited EMT via blocking the TGF-β1/SMAD pathway, which suppressed GC cell growth and liver metastasis. This study provides data to support the treatment of liver metastasis in GC with TCM and holds significant importance for the research and development of new anticancer drugs.
Collapse
Affiliation(s)
- Wenjing Liu
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Doudou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Shumin Jia
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yating Yang
- The Second Hospital of Chinese Medicine of BAO JI City, Baoji, 721300, Xian, China
| | - Fandi Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Yang
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Nan
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
3
|
Wu H, Zhu X, Zhou H, Sha M, Ye J, Yu H. Mitochondrial Ribosomal Proteins and Cancer. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:96. [PMID: 39859078 PMCID: PMC11766452 DOI: 10.3390/medicina61010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025]
Abstract
Mitochondria play key roles in maintaining cell life and cell function, and their dysfunction can lead to cell damage. Mitochondrial ribosomal proteins (MRPs) are encoded by nuclear genes and are assembled within the mitochondria. MRPs are pivotal components of the mitochondrial ribosomes, which are responsible for translating 13 mitochondrial DNA-encoded proteins essential for the mitochondrial respiratory chain. Recent studies have underscored the importance of MRPs in cancer biology, revealing their altered expression patterns in various types of cancer and their potential as both prognostic biomarkers and therapeutic targets. Herein, we review the current knowledge regarding the multiple functions of MRPs in maintaining the structure of the mitochondrial ribosome and apoptosis, their implications for cancer susceptibility and progression, and the innovative strategies being developed to target MRPs and mitoribosome biogenesis in cancer therapy. This comprehensive overview aims to provide insights into the role of MRPs in cancer biology and highlight promising strategies for future precision oncology.
Collapse
Affiliation(s)
- Huiyi Wu
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China; (H.W.); (X.Z.); (H.Z.)
| | - Xiaowei Zhu
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China; (H.W.); (X.Z.); (H.Z.)
| | - Huilin Zhou
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China; (H.W.); (X.Z.); (H.Z.)
| | - Min Sha
- Translational Medicine Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China; (M.S.); (J.Y.)
| | - Jun Ye
- Translational Medicine Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China; (M.S.); (J.Y.)
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China; (H.W.); (X.Z.); (H.Z.)
- Translational Medicine Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China; (M.S.); (J.Y.)
| |
Collapse
|
4
|
Bacon JM, Jones JL, Liu GS, Dickinson JL, Raspin K. Mitochondrial ribosomal proteins in metastasis and their potential use as prognostic and therapeutic targets. Cancer Metastasis Rev 2024; 43:1119-1135. [PMID: 39354291 PMCID: PMC11554709 DOI: 10.1007/s10555-024-10216-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
The mitochondrion is an essential cell organelle known as the powerhouse of the cell. Mitochondrial ribosomal proteins (MRPs) are nuclear encoded, synthesised in the cytoplasm but perform their main functions in the mitochondria, which includes translation, transcription, cell death and maintenance. However, MRPs have also been implicated in cancer, particularly advanced disease and metastasis across a broad range of cancer types, where they play a central role in cell survival and progression. For some, their altered expression has been investigated as potential prognostic markers, and/or therapeutic targets, which is the focus of this review. Several therapies targeting MRPs are currently approved by the Food and Drug Administration and the European Medicines Agency for use in other diseases, revealing the opportunity for repurposing their use in advanced and metastatic cancer. Herein, we review the evidence supporting key MRPs as molecular drivers of advanced disease in multiple cancer types. We also highlight promising avenues for future use of MRPs as precision targets in the treatment of late-stage cancers for which there are currently very limited effective treatment options.
Collapse
Affiliation(s)
- Jasmine M Bacon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Johanna L Jones
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Guei-Sheung Liu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, Victoria, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kelsie Raspin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| |
Collapse
|
5
|
Guo H, Dong Y, Luo D, Gong M, Sun J, Wu Z, Liu Z, Zhong L, Jin S. MRPL41, as a target for acupuncture, promotes neuron apoptosis in models of ischemic stroke via activating p53 pathway. Neurochem Int 2024; 180:105881. [PMID: 39406282 DOI: 10.1016/j.neuint.2024.105881] [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: 07/04/2024] [Revised: 09/24/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
Neuronal death is the key cause of ischemic stroke. Acupuncture (Acu) is a recognized method for the treatment and amelioration of cerebral ischemia. However, the molecular mechanism of Acu for treating ischemic stroke has not yet been detailedly elucidated. Based our microarray analysis results, mitochondrial ribosomal protein L41 (MRPL41), which is related to apoptosis, was identified as the target of Acu. MRPL41 expression was increased in middle cerebral artery occlusion/reperfusion (MCAO/R) model and reduced after Acu treatment. Following, MCAO/R model and oxygen and glucose deprivation/reoxygenation (OGD/R) model were established to explore the effect of MRPL41. Knockdown of MRPL41 increased cell viability and ani-apoptotic protein (Bcl-2) expression, and reduced apoptosis intensity and pro-apoptotic protein (Bax and Cleaved caspase-3) of OGD/R neurons. In vivo, MRPL41 silencing decreased neurological severity score, shrank infarct area, reduced encephaledema and neuron apoptosis. In addition, reduction of MRPL41 caused loss of p53. Our data uncover that Acu targets MRPL41, following with inhibiting neuron apoptosis via p53 pathway, thereby ameliorating ischemic stroke.
Collapse
Affiliation(s)
- Hong Guo
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanwei Dong
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Danqing Luo
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meng Gong
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianfeng Sun
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhipeng Wu
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhixiang Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lei Zhong
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Song Jin
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
6
|
Chen N, Zhou H, He B, Peng S, Ding F, Liu QH, Ma Z, Liu W, Xu B. Melatonin promotes cell cycle progression of neural stem cells subjected to manganese via Nurr1. ENVIRONMENTAL TOXICOLOGY 2024; 39:3883-3896. [PMID: 38563506 DOI: 10.1002/tox.24258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/04/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
Excessive exposure to manganese (Mn) through drinking water and food during pregnancy significantly heightens the likelihood of neurodevelopmental damage in offspring. Multiple studies have indicated that melatonin (Mel) may help to relieve neurodevelopmental disorders caused by Mn, but potential mechanisms underlying this effect require further exploration. Here, we utilized primary neural stem cells (NSCs) as a model to elucidate the molecular mechanism underlying the protective function of Mel on Mn-induced cell proliferation dysfunction and cycle arrest. Our results showed that Mn disrupted the cell cycle in NSCs by suppressing positive regulatory proteins (CDK2, Cyclin A, Cyclin D1, and E2F1) and enhancing negative ones (p27KIP1 and p57KIP2), leading to cell proliferation dysfunction. Mel inhibited the Mn-dependent changes to these proteins and the cell cycle through nuclear receptor-related protein 1 (Nurr1), thus alleviating the proliferation dysfunction. Knockdown of Nurr1 using lentivirus-expressed shRNA in NSCs resulted in a diminished protective effect of Mel. We concluded that Mel mitigated Mn-induced proliferation dysfunction and cycle arrest in NSCs through Nurr1.
Collapse
Affiliation(s)
- Nan Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Han Zhou
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Bin He
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Sen Peng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Feng Ding
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Qi-Hao Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Zhuo Ma
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
- Key laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, PR China
| |
Collapse
|
7
|
Yang Y, Yuan L, Meng F, Lu D, Che M, Zhou X, Chen G, Ning N, Nan Y. Gancao Xiexin Decoction inhibits gastric carcinoma proliferation and migration by regulating the JAK2/STAT3 signalling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117241. [PMID: 37777026 DOI: 10.1016/j.jep.2023.117241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/18/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The incidence of gastric carcinoma (GC) is increasing rapidly. Traditional Chinese Medicine (TCM) plays a unique role in the treatment of GC. At present, Gancao Xiexin Decoction (GCXXD) has been proved to have a good therapeutic effect on diseases of the spleen and stomach system, but relevant molecular mechanisms remain incompletely explained. AIM OF STUDY The mechanism of GCXXD for GC was investigated by network pharmacology and verified by cell experiments. MATERIALS AND METHODS Firstly, the public database was used to identify the core targets and key pathways of GCXXD in treating GC, followed by molecular docking and survival analysis. Subsequently, the effects of GCXXD on human gastric cancer AGS and HGC-27 cells were confirmed by a series of experiments, such as CCK-8, colony formation, apoptosis, cell cycle, wound scratch assay, transwell chamber assay, qRT-PCR and Western blot. RESULTS This study identified quercetin, wogonin, kaempferol, baicalein, sitosterol and beta-sitosterol as key ingredients, along with AKT1, TP53, JUN, STAT3, TNF, MAPK3, HSP90AA1 and EGFR as co targets, and the JAK/STAT signalling pathway as the key pathway. The experimental results showed that GCXXD inhibited the growth of GC cells, increased the apoptosis rate and the ratio of G0/G1 phase cells, and weakened the clone formation rate and inhibited cell migration and invasion. It also reduces the expression of core target genes and downregulates the expression of JAK2, p-JAK2, STAT3, and p-STAT3 proteins. CONCLUSION GCXXD inhibits GC cell growth, reduces clonogenic capacity, induces apoptosis, blocks the cell cycle, and decreases cell migration and invasion rates by inhibiting the JAK2/STAT3 signalling pathway.
Collapse
Affiliation(s)
- Yating Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Fandi Meng
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Doudou Lu
- Clinical Medical College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous, China
| | - Mengying Che
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Xin Zhou
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous, China
| | - Guoqing Chen
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Na Ning
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous, China
| | - Yi Nan
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
8
|
Du YH, Zhao JJ, Li X, Huang SC, Ning N, Chen GQ, Yang Y, Nan Y, Yuan L. Mechanism of pachymic acid in the treatment of gastric cancer based on network pharmacology and experimental verification. World J Gastrointest Oncol 2024; 16:30-50. [PMID: 38292852 PMCID: PMC10824110 DOI: 10.4251/wjgo.v16.i1.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/07/2023] [Accepted: 11/02/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Pachymic acid (PA) is derived from Poria cocos. PA has a variety of pharmacological and inhibitory effects on various tumors. However, the mechanism of action of PA in gastric cancer (GC) remains unclear. AIM To investigate the mechanism of PA in treating GC via the combination of network pharmacology and experimental verification. METHODS The GeneCards and OMIM databases were used to derive the GC targets, while the Pharm Mapper database provided the PA targets. Utilizing the STRING database, a protein-protein interaction network was constructed and core targets were screened. The analyses of Gene Ontology, Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis were conducted, and molecular docking and clinical correlation analyses were performed on the core targets. Ultimately, the network pharmacology findings were validated through in vitro cell assays, encompassing assessments of cell viability, apoptosis, cell cycle, cloning, and western blot analysis. RESULTS According to network pharmacology analysis, the core targets were screened, and the PI3K/AKT signaling pathway is likely to be the mechanism by which PA effectively treats GC, according to KEGG enrichment analysis. The experimental findings showed that PA could control PI3K/AKT signaling to prevent GC cell proliferation, induce apoptosis, and pause the cell cycle. CONCLUSION Network pharmacology demonstrated that PA could treat GC by controlling a variety of signaling pathways and acting on a variety of targets. This has also been supported by in vitro cell studies, which serve as benchmarks for further research.
Collapse
Affiliation(s)
- Yu-Hua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jian-Jun Zhao
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Xia Li
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Ningxia Chinese Medicine Research Center, Manufacturing Laboratory, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shi-Cong Huang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Na Ning
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Guo-Qing Chen
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Yang
- College of Foundation, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
9
|
Yang Y, Nan Y, Du YH, Huang SC, Lu DD, Zhang JF, Li X, Chen Y, Zhang L, Yuan L. 18β-glycyrrhetinic acid promotes gastric cancer cell autophagy and inhibits proliferation by regulating miR-328-3p/signal transducer and activator of transcription 3. World J Gastroenterol 2023; 29:4317-4333. [PMID: 37545635 PMCID: PMC10401664 DOI: 10.3748/wjg.v29.i27.4317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/10/2023] [Accepted: 06/02/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common cancer types worldwide, and its prevention and treatment methods have garnered much attention. As the active ingredient of licorice, 18β-glycyrrhetinic acid (18β-GRA) has a variety of pharmacological effects. The aim of this study was to explore the effective target of 18β-GRA in the treatment of GC, in order to provide effective ideas for the clinical prevention and treatment of GC. AIM To investigate the mechanism of 18β-GRA in inhibiting cell proliferation and promoting autophagy flux in GC cells. METHODS Whole transcriptomic analyses were used to analyze and screen differentially expressed microRNAs (miRNAs) in GC cells after 18β-GRA intervention. Lentivirus-transfected GC cells and the Cell Counting Kit-8 were used to detect cell proliferation ability, cell colony formation ability was detected by the clone formation assay, and flow cytometry was used to detect the cell cycle and apoptosis. A nude mouse transplantation tumor model of GC cells was constructed to verify the effect of miR-328-3p overexpression on the tumorigenicity of GC cells. Tumor tissue morphology was observed by hematoxylin and eosin staining, and microtubule-associated protein light chain 3 (LC3) expression was detected by immunohistochemistry. TransmiR, STRING, and miRWalk databases were used to predict the relationship between miR-328-3p and signal transducer and activator of transcription 3 (STAT3)-related information. Expression of STAT3 mRNA and miR-328-3p was detected by quantitative polymerase chain reaction (qPCR) and the expression levels of STAT3, phosphorylated STAT3 (p-STAT3), and LC3 were detected by western blot analysis. The targeted relationship between miR-328-3p and STAT3 was detected using the dual-luciferase reporter gene system. AGS cells were infected with monomeric red fluorescent protein-green fluorescent protein-LC3 adenovirus double label. LC3 was labeled and autophagy flow was observed under a confocal laser microscope. RESULTS The expression of miR-328-3p was significantly upregulated after 18β-GRA intervention in AGS cells (P = 4.51E-06). Overexpression of miR-328-3p inhibited GC cell proliferation and colony formation ability, arrested the cell cycle in the G0/G1 phase, promoted cell apoptosis, and inhibited the growth of subcutaneous tumors in BALB/c nude mice (P < 0.01). No obvious necrosis was observed in the tumor tissue in the negative control group (no drug intervention or lentivirus transfection) and vector group (the blank vector for lentivirus transfection), and more cells were loose and necrotic in the miR-328-3p group. Bioinformatics tools predicted that miR-328-3p has a targeting relationship with STAT3, and STAT3 was closely related to autophagy markers such as p62. After overexpressing miR-328-3p, the expression level of STAT3 mRNA was significantly decreased (P < 0.01) and p-STAT3 was downregulated (P < 0.05). The dual-luciferase reporter gene assay showed that the luciferase activity of miR-328-3p and STAT3 3' untranslated regions of the wild-type reporter vector group was significantly decreased (P < 0.001). Overexpressed miR-328-3p combined with bafilomycin A1 (Baf A1) was used to detect the expression of LC3 II. Compared with the vector group, the expression level of LC3 II in the overexpressed miR-328-3p group was downregulated (P < 0.05), and compared with the Baf A1 group, the expression level of LC3 II in the overexpressed miR-328-3p + Baf A1 group was upregulated (P < 0.01). The expression of LC3 II was detected after intervention of 18β-GRA in GC cells, and the results were consistent with the results of miR-328-3p overexpression (P < 0.05). Additional studies showed that 18β-GRA promoted autophagy flow by promoting autophagosome synthesis (P < 0.001). qPCR showed that the expression of STAT3 mRNA was downregulated after drug intervention (P < 0.05). Western blot analysis showed that the expression levels of STAT3 and p-STAT3 were significantly downregulated after drug intervention (P < 0.05). CONCLUSION 18β-GRA promotes the synthesis of autophagosomes and inhibits GC cell proliferation by regulating the miR-328-3p/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yi Yang
- Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yu-Hua Du
- Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shi-Cong Huang
- Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Dou-Dou Lu
- Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jun-Fei Zhang
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Xia Li
- Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yan Chen
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Lei Zhang
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
10
|
Zong L, Wang Y, Song S, Zhang H, Mu S, Liu W, Feng Y, Wang S, Tu Z, Yuan Q, Li L, Pu X. Formulation and Evaluation on Synergetic Anti-Hepatoma Effect of a Chemically Stable and Release-Controlled Nanoself-Assembly with Natural Monomers. Int J Nanomedicine 2023; 18:3407-3428. [PMID: 37377983 PMCID: PMC10292624 DOI: 10.2147/ijn.s408416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction Hepatoma is the leading cause of death among liver diseases worldwide. Modern pharmacological studies suggest that some natural monomeric compounds have a significant effect on inhibiting tumor growth. However, poor stability and solubility, and side effects are the main factors limiting the clinical application of natural monomeric compounds. Methods In this paper, drug-co-loaded nanoself-assemblies were selected as a delivery system to improve the chemical stability and solubility of Tanshinone II A and Glycyrrhetinic acid, and to produce a synergetic anti-hepatoma effect. Results The study suggested that the drug co-loaded nanoself-assemblies showed high drug loading capacity, good physical and chemical stability, and controlled release. In vitro cell experiments verified that the drug-co-loaded nanoself-assemblies could increase the cellular uptake and cell inhibitory activity. In vivo studies verified that the drug co-loaded nanoself-assemblies could prolong the MRT0-∞, increase accumulation in tumor and liver tissues, and show strong synergistic anti-tumor effect and good bio-safety in H22 tumor-bearing mice. Conclusion This work indicates that natural monomeric compounds co-loaded nanoself-assemblies would be a potential strategy for the treatment of hepatoma.
Collapse
Affiliation(s)
- Lanlan Zong
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Yanling Wang
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Shiyu Song
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Huiqi Zhang
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Shengcai Mu
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Wenshang Liu
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Yu Feng
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Shumin Wang
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Ziwei Tu
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Qi Yuan
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| | - Luhui Li
- Medical school, Henan Technical Institute, Kaifeng, Henan, 475004, People’s Republic of China
| | - Xiaohui Pu
- School of Pharmacy and Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| |
Collapse
|
11
|
Li X, Ma XL, Nan Y, Du YH, Yang Y, Lu DD, Zhang JF, Chen Y, Zhang L, Niu Y, Yuan L. 18β-glycyrrhetinic acid inhibits proliferation of gastric cancer cells through regulating the miR-345-5p/TGM2 signaling pathway. World J Gastroenterol 2023; 29:3622-3644. [PMID: 37398884 PMCID: PMC10311615 DOI: 10.3748/wjg.v29.i23.3622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/24/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common gastrointestinal malignancy worldwide. Based on cancer-related mortality, the current prevention and treatment strategies for GC still show poor clinical results. Therefore, it is important to find effective drug treatment targets.
AIM To explore the molecular mechanism of 18β-glycyrrhetinic acid (18β-GRA) regulating the miR-345-5p/TGM2 signaling pathway to inhibit the proliferation of GC cells.
METHODS CCK-8 assay was used to determine the effect of 18β-GRA on the survival rate of GES-1 cells and AGS and HGC-27 cells. Cell cycle and apoptosis were detected by flow cytometry, cell migration was detected by a wound healing assay, the effect of 18β-GRA on subcutaneous tumor growth in BALB/c nude mice was investigated, and the cell autophagy level was determined by MDC staining. TMT proteomic analysis was used to detect the differentially expressed autophagy-related proteins in GC cells after 18β-GRA intervention, and then the protein-protein interaction was predicted using STRING (https://string-db.org/). MicroRNAs (miRNAs) transcriptome analysis was used to detect the miRNA differential expression profile, and use miRBase (https://www.mirbase/) and TargetScan (https://www.targetscan.org/) to predict the miRNA and complementary binding sites. Quantitative real-time polymerase chain reaction was used to detect the expression level of miRNA in 18β-GRA treated cells, and western blot was used to detect the expression of autophagy related proteins. Finally, the effect of miR-345-5p on GC cells was verified by mir-345-5p overexpression.
RESULTS 18β-GRA could inhibit GC cells viability, promote cell apoptosis, block cell cycle, reduce cell wound healing ability, and inhibit the GC cells growth in vivo. MDC staining results showed that 18β-GRA could promote autophagy in GC cells. By TMT proteomic analysis and miRNAs transcriptome analysis, it was concluded that 18β-GRA could down-regulate TGM2 expression and up-regulate miR-345-5p expression in GC cells. Subsequently, we verified that TGM2 is the target of miR-345-5p, and that overexpression of miR-345-5p significantly inhibited the protein expression level of TGM2. Western blot showed that the expression of autophagy-related proteins of TGM2 and p62 was significantly reduced, and LC3II, ULK1 and AMPK expression was significantly increased in GC cells treated with 18β-GRA. Overexpression of miR-345-5p not only inhibited the expression of TGM2, but also inhibited the proliferation of GC cells by promoting cell apoptosis and arresting cell cycle.
CONCLUSION 18β-GRA inhibits the proliferation of GC cells and promotes autophagy by regulating the miR-345-5p/TGM2 signaling pathway.
Collapse
Affiliation(s)
- Xia Li
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Xiao-Ling Ma
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yu-Hua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Yang
- College of Basic Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Dou-Dou Lu
- College of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jun-Fei Zhang
- College of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yan Chen
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Lei Zhang
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yang Niu
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
12
|
Li X, Du Y, Huang S, Yang Y, Lu D, Zhang J, Chen Y, Zhang L, Nan Y, Yuan L. Exploring the molecular mechanism of glycyrrhetinic acid in the treatment of gastric cancer based on network pharmacology and experimental validation. Aging (Albany NY) 2023; 15:3839-3856. [PMID: 37171392 PMCID: PMC10449304 DOI: 10.18632/aging.204718] [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: 02/13/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023]
Abstract
There is a wide range of pharmacological effects for glycyrrhetinic acid (GRA). Previous studies have shown that GRA could inhibit the proliferation of tumor cells, showing a promising value in the treatment of gastric cancer (GC). Nonetheless, the precise mechanism of the effect of GRA on GC remains unclear. We explored cellular and molecular mechanisms of GRA based on network pharmacology and in vitro experimental validation. In this study, we predicted 156 potential therapeutic targets for GC with GRA from public databases. We then screened the hub targets using protein-protein interaction network (PPI) and conducted clinical correlation analysis. Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment showed that GRA made anti-GC effects through multiple targets and pathways, particularly the MAPK signaling pathway. Next, molecular docking results revealed a potential interaction between GRA and MAPK3. In addition, qRT-PCR experiments revealed that 18β-GRA was able to suppress mRNA expression of KRAS, ERK1 and ERK2 in AGS cells. Western blotting results also revealed that 18β-GRA was able to suppress the expression of KRAS and p-ERK1/2 proteins in AGS cells. Additionally, immunofluorescence assays revealed that 18β-GRA inhibited p-ERK1/2 nuclear translocation in AGS cells. These results systematically reveal that 18β-GRA may have anti-tumor effects on GC by modulating the MAPK signaling pathway.
Collapse
Affiliation(s)
- Xia Li
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shicong Huang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Yang
- College of Basic Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Doudou Lu
- College of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Junfei Zhang
- College of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yan Chen
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Lei Zhang
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|