1
|
Huang W, Wen F, Yang P, Li Y, Li Q, Shu P. Yi-qi-hua-yu-jie-du decoction induces ferroptosis in cisplatin-resistant gastric cancer via the AKT/GSK3β/NRF2/GPX4 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155220. [PMID: 38056149 DOI: 10.1016/j.phymed.2023.155220] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/28/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Resistance to chemotherapy in gastric cancer (GC) is a ubiquitous challenge for its treatment. Yi-qi-hua-yu-jie-du decoction (YJD), an empirical formula in Traditional Chinese Medicine (TCM), demonstrated survival-prolonging functions in patients with GC. Previous research has shown that YJD could also inhibit drug resistance in GC. However, the precise mechanisms for how YJD accomplishes this remain incompletely explained. PURPOSE The research aimed to identify differential metabolic characteristics in cisplatin-resistant GC and investigate whether YJD can target these differences to suppress GC drug resistance. METHODS Metabolomic analysis was conducted to identify metabolic disparities between cisplatin-resistant and parental GC cells, as well as metabolic modifications resulting from YJD intervention in cisplatin-resistant GC cells. The effect of YJD on ferroptosis stimulation was assessed by measuring the levels of reactive oxygen species (ROS), malondialdehyde (MDA), iron ions, the reduced glutathione (GSH) to oxidised glutathione (GSSG) ratio, and alterations in mitochondrial morphology. Western blotting and quantitative real-time polymerase chain reaction (Q-PCR) were employed to verity the mechanisms of YJD-triggered ferroptosis through GPX4 and NRF2 overexpression models, alongside the AKT activator SC79. In vivo validation was conducted using nude mouse xenograft models. RESULTS Cisplatin-resistant GC exhibited altered GSH/GPX4 metabolism, and ferroptosis was a significantly enriched cell death pattern with YJD treatment in cisplatin-resistant GC cells. Ferroptosis biomarkers, including ROS, MDA, iron ions, the GSH/GSSG ratio, and mitochondrial morphology, were remarkably changed with the YJD intervention. Mechanistic experiments demonstrated that YJD inhibited the phosphorylation cascade activity of the AKT/GSK3β pathway, thereby reducing NRF2 expression. The level of GPX4, a crucial enzyme involved in glutathione metabolism, was attenuated, facilitating ferroptosis induction in cisplatin-resistant GC. CONCLUSION The research reveals, for the first time, changes in GSH/GPX4 metabolism in cisplatin-resistant GC cells based on metabolomic analysis. YJD induced ferroptosis in cisplatin-resistant GC by inhibiting GPX4 through the AKT/GSK3β/NRF2 pathway, thus attenuating the cisplatin drug resistance in GC. Our findings identify metabolic changes in cisplatin-resistant GC and establish a theoretical framework for YJD on tackling drug resistance in GC through ferroptosis.
Collapse
Affiliation(s)
- Wenjie Huang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Fang Wen
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Peipei Yang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Ye Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Qiurong Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Peng Shu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China; School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
2
|
Zhang Y, Shi D, Zhang X, Wu S, Liu W, Luo B. Downregulation of MUS81 expression inhibits cell migration and maintains EBV latent infection through miR-BART9-5p in EBV-associated gastric cancer. J Med Virol 2023; 95:e28725. [PMID: 37185865 DOI: 10.1002/jmv.28725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/15/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023]
Abstract
Epstein-Barr virus (EBV) infection is associated with the occurrence and development of gastric cancer (GC). Methyl methanesulfonate and ultraviolet-sensitive gene 81 (MUS81) is the catalytic component of a structure-specific endonuclease and plays an important role in chromosomal stability. However, the link between EBV infection and MUS81 remains unclear. In the present study, we found that MUS81 expression was much lower in EBV-associated GC cells than in EBV-negative GC. MUS81 acts as an oncogene in GC by inducing the cell migration and proliferation. Western blot and luciferase reporter assays revealed that miR-BART9-5p directly targeted MUS81 and downregulated its expression. Additionally, overexpression of MUS81 in EBV-positive GC cells inhibited the expression of EBV nuclear antigen 1 (EBNA1). EBNA1 is critical for the pathogenesis of EBV-associated tumors and the maintenance of a stable copy number of the viral genomes. Altogether, these results indicated that the lowering MUS81 expression might be a mechanism by EBV to maintain its latent infection.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathogeny Biology, Basic Medicine College, Qingdao University, Qingdao, China
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Duo Shi
- Department of Pathogeny Biology, Basic Medicine College, Qingdao University, Qingdao, China
| | - Xing Zhang
- Department of Pathogeny Biology, Basic Medicine College, Qingdao University, Qingdao, China
| | - Shuo Wu
- Department of Pathogeny Biology, Basic Medicine College, Qingdao University, Qingdao, China
- Laboratory Medicine Center of Qingdao, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Wen Liu
- Department of Pathogeny Biology, Basic Medicine College, Qingdao University, Qingdao, China
| | - Bing Luo
- Department of Pathogeny Biology, Basic Medicine College, Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Lin X, Yang X, Yang Y, Zhang H, Huang X. Research progress of traditional Chinese medicine as sensitizer in reversing chemoresistance of colorectal cancer. Front Oncol 2023; 13:1132141. [PMID: 36994201 PMCID: PMC10040588 DOI: 10.3389/fonc.2023.1132141] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/27/2023] [Indexed: 03/14/2023] Open
Abstract
In recent years, the incidences and mortalities from colorectal cancer (CRC) have been increasing; therefore, there is an urgent need to discover newer drugs that enhance drug sensitivity and reverse drug tolerance in CRC treatment. With this view, the current study focuses on understanding the mechanism of CRC chemoresistance to the drug as well as exploring the potential of different traditional Chinese medicine (TCM) in restoring the sensitivity of CRC to chemotherapeutic drugs. Moreover, the mechanism involved in restoring sensitivity, such as by acting on the target of traditional chemical drugs, assisting drug activation, increasing intracellular accumulation of anticancer drugs, improving tumor microenvironment, relieving immunosuppression, and erasing reversible modification like methylation, have been thoroughly discussed. Furthermore, the effect of TCM along with anticancer drugs in reducing toxicity, increasing efficiency, mediating new ways of cell death, and effectively blocking the drug resistance mechanism has been studied. We aimed to explore the potential of TCM as a sensitizer of anti-CRC drugs for the development of a new natural, less-toxic, and highly effective sensitizer to CRC chemoresistance.
Collapse
Affiliation(s)
- Xiang Lin
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyu Yang
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yushang Yang
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hangbin Zhang
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuan Huang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Xuan Huang,
| |
Collapse
|
4
|
Huang W, Wen F, Ruan S, Gu P, Gu S, Song S, Zhou J, Li Y, Liu J, Shu P. Integrating HPLC-Q-TOF-MS/MS, network pharmacology and experimental validation to decipher the chemical substances and mechanism of modified Gui-shao-liu-jun-zi decoction against gastric cancer. J Tradit Complement Med 2023; 13:245-262. [PMID: 37128200 PMCID: PMC10148141 DOI: 10.1016/j.jtcme.2023.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 12/17/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Background and aim Gastric cancer (GC) is a common malignant tumor worldwide. Modified Gui-shao-liu-jun-zi decoction (mGSLJZ) is a clinically effective traditional Chinese medicine (TCM) compound in GC treatment. This study aimed to analyze main chemical substances of mGSLJZ and investigate active ingredients and molecular mechanism of mGSLJZ against GC. Experimental procedure HPLC-Q-TOF-MS/MS was used to analyze chemical substances of mGSLJZ, and potential active ingredients were screened from TCMSP. The target set of mGSLJZ for GC was obtained based on SwissTargetPrediction. The PPI network was constructed to screen out core targets. GO and KEGG enrichment analyses were conducted to identify BPs, CCs, MFs and pathways. The "active ingredient-core target-pathway" regulatory network was constructed to obtain core substances. Subsequently, Oncomine, Proteinatlas and molecular docking were performed to validate these findings. The cell experiments were conducted to confirm the anti-GC effects of mGLSJZ. Results and conclusion Forty-one potential active ingredients were filtered out from 120 chemical substances in mGSLJZ, including various organic acids and flavonoids. The top 10 key targets, 20 related pathways and 6 core medicinal substances were obtained based on network pharmacology analysis. Molecular docking results indicated that the core substances and key targets had good binding activities. The cell experiments validated that mGSLJZ and the core substances inhibited the proliferation in multiple GC cells and that mGLSJZ restrained the migration of GC. Meanwhile, the top 5 targets and top 2 pathways were verified. The rescue experiments demonstrated that mGSLJZ suppressed the proliferation and migration of GC through the PI3K/AKT/HIF-1 pathway.
Collapse
Affiliation(s)
- Wenjie Huang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Fang Wen
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuai Ruan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Peixing Gu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Suping Gu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyuan Song
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiayu Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiatong Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Shu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
- Corresponding author. 155 Hanzhong Road, Nanjing, Jiangsu Province, 210000, China.
| |
Collapse
|
5
|
Huang W, Wen F, Gu P, Liu J, Xia Y, Li Y, Zhou J, Song S, Ruan S, Gu S, Chen X, Shu P. The inhibitory effect and mechanism of Yi-qi-hua-yu-jie-du decoction on the drug resistance of gastric cancer stem cells based on ABC transporters. Chin Med 2022; 17:93. [PMID: 35941687 PMCID: PMC9361523 DOI: 10.1186/s13020-022-00647-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/29/2022] [Indexed: 01/12/2023] Open
Abstract
Background The drug resistance of tumor stem cells is an obstacle in gastric cancer (GC) treatment and the high expression of ABC transporters is a classic reason for drug resistance. This study aimed to construct a reliable GC drug-resistant stem cell model and explore the inhibitory effect and mechanism of Yi-qi-hua-yu-jie-du medicated serum (YQHY) on the drug resistance of GC stem cells based on ABC transporters. Methods The tumor stemness biomarker CD44 was primary identification from WGCNA. The magnetic-activated cell sorting (MACS) method was used to separate CD44( +)BGC823/5-Fu (BGC823/5–Fu-CSCs) cells and the stemness characteristics were verified from multiple dimensions. Then, the drug resistance index and expression of ABC transporter genes MDR1 and MRP1 were detected in CD44(−)/CD44(+) cells. The inhibition and apoptosis rates of the cells administrated with YQHY or/and 5-Fu were calculated to confirm that YQHY can suppress the drug resistance of BGC823/5-Fu-CSCs. Afterwards, the effects of YQHY on the expression of MDR1 and MRP1 and the activation of the PI3K/Akt/Nrf2 pathway were observed. Finally, under the administration of IGF-1 (the activator of PI3K/Akt pathway) and Nrf2 siRNA, the mechanism of YQHY on reversing the drug resistance of BGC823/5–Fu-CSCs through inhibiting the expression of MDR1 and MRP1 via PI3K/Akt/Nrf2 was verified. Results CD44 was a reliable GC stemness biomarker and can be applied to construct the drug-resistant GC stem cell model CD44(+)BGC823/5-Fu. The growth rate, cell proliferation index, soft agar colony formation, expression of stemness specific genes and tumorigenesis ability of CD44(+)BGC823/5-Fu cells were significantly higher than those of CD44(−)BGC823/5-Fu cells. BGC823/5–Fu-CSCs exhibited strong drug resistance to 5-Fu and high expression of ABC transporter genes MDR1 and MRP1 compared to CD44(-) cells. YQHY increased the inhibition and apoptosis rates to efficiently inhibit the drug resistance of BGC823/5–Fu-CSCs. Meanwhile, it suppressed the expression of MDR1 and MRP1 and restrained the activation of PI3K/Akt/Nrf2 signaling pathway. Finally, it was found that IGF-1 partially restored the activation of PI3K/Akt/Nrf2 pathway, alleviated the inhibition of MDR1 and MRP1, blocked the proliferation-inhibitory and apoptosis-promotion effects. YQHY and si-Nrf2 synergistically suppressed the MDR1/MRP1 expression and the drug resistance of BGC823/5–Fu-CSCs. Conclusions CD44 was a reliable GC stemness biomarker, and the high expression of ABC transporter genes MDR1 and MRP1 was an important feature of drug-resistant stem cells. YQHY inhibited the MDR1 and MRP1 expression via PI3K/Akt/Nrf2 pathway, thus reversing the drug resistance of BGC823/5–Fu-CSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-022-00647-y.
Collapse
Affiliation(s)
- Wenjie Huang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fang Wen
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peixing Gu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiatong Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Xia
- Department of Respiratory, Wujin Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Ye Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiayu Zhou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyuan Song
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuai Ruan
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Suping Gu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoxue Chen
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China.,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Shu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210000, Jiangsu, China. .,First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
6
|
Jangili P, Kong N, Kim JH, Zhou J, Liu H, Zhang X, Tao W, Kim JS. DNA-Damage-Response-Targeting Mitochondria-Activated Multifunctional Prodrug Strategy for Self-Defensive Tumor Therapy. Angew Chem Int Ed Engl 2022; 61:e202117075. [PMID: 35133703 DOI: 10.1002/anie.202117075] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Indexed: 12/14/2022]
Abstract
We report a novel multifunctional construct, M1, designed explicitly to target the DNA damage response in cancer cells. M1 contains both a floxuridine (FUDR) and protein phosphatase 2A (PP2A) inhibitor combined with a GSH-sensitive linker. Further conjugation of the triphenylphosphonium moiety allows M1 to undergo specific activation in the mitochondria, where mitochondria-mediated apoptosis is observed. Moreover, M1 has enormous effects on genomic DNA ascribed to FUDR's primary function of impeding DNA/RNA synthesis combined with diminishing PP2A-activated DNA repair pathways. Importantly, mechanistic studies highlight the PP2A obtrusion in FUDR/5-fluorouracil (5-FU) therapy and underscore the importance of its inhibition to harbor therapeutic potential. HCT116 cell xenograft-bearing mice that have a low response rate to 5-FU show a prominent effect with M1, emphasizing the importance of DNA damage response targeting strategies using tumor-specific microenvironment-activatable systems.
Collapse
Affiliation(s)
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, 311121, China.,Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ji Hyeon Kim
- Department of Chemistry, Korea University, Seoul, 02841, Korea
| | - Jun Zhou
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Haijun Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xingcai Zhang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02134, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, Korea
| |
Collapse
|
7
|
Jangili P, Kong N, Kim JH, Zhou J, Liu H, Zhang X, Tao W, Kim JS. DNA‐Damage‐Response‐Targeting Mitochondria‐Activated Multifunctional Prodrug Strategy for Self‐Defensive Tumor Therapy. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202117075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | - Na Kong
- Liangzhu Laboratory Zhejiang University Medical Center Hangzhou Zhejiang 311121 China
- Center for Nanomedicine and Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Ji Hyeon Kim
- Department of Chemistry Korea University Seoul 02841 Korea
| | - Jun Zhou
- Center for Nanomedicine and Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Haijun Liu
- Center for Nanomedicine and Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Xingcai Zhang
- School of Engineering and Applied Sciences Harvard University Cambridge MA 02134 USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Jong Seung Kim
- Department of Chemistry Korea University Seoul 02841 Korea
| |
Collapse
|
8
|
Song S, Wen F, Gu S, Gu P, Huang W, Ruan S, Chen X, Zhou J, Li Y, Liu J, Shu P. Network Pharmacology Study and Experimental Validation of Yiqi Huayu Decoction Inducing Ferroptosis in Gastric Cancer. Front Oncol 2022; 12:820059. [PMID: 35237519 PMCID: PMC8883049 DOI: 10.3389/fonc.2022.820059] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
ObjectiveThis study aimed to identify the mechanism of Yiqi Huayu Decoction (YQHY) induced ferroptosis in gastric cancer (GC) by using network pharmacology and experimental validation.MethodsThe targets of YQHY, ferroptosis-related targets, and targets related to GC were derived from databases. Following the protein–protein interaction (PPI) network, the hub targets for YQHY induced ferroptosis in GC were identified. Furthermore, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment were used to analyze the hub targets from a macro perspective. We verified the hub targets by molecular docking, GEPIA, HPA, and the cBioPortal database. Finally, we performed cell viability assays, quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, lipid peroxidation, and GSH assays to explore the mechanism of YQHY induced ferroptosis in GC.ResultsWe identified the main active compounds and hub targets: Quercetin, DIBP, DBP, Mipax, Phaseol and TP53, ATM, SMAD4, PTGS2, and ACSL4. KEGG enrichment analyses indicated that the JAK2-STAT3 signaling pathway may be a significant pathway. Molecular docking results showed that the main active compounds had a good binding activity with the hub targets. The experimental results proved that YQHY could induce ferroptosis in AGS by increasing the MDA content and reducing the GSH content. qRT–PCR and Western blot results showed that YQHY can induce ferroptosis in GC by affecting the JAK2-STAT3 pathway and the expression of ACSL4.ConclusionsThis study indicated that YQHY can induce ferroptosis in GC by affecting the JAK2–STAT3 pathway and the expression of ACSL4, and induction of ferroptosis may be one of the possible mechanisms of YQHY’s anti-recurrence and metastasis of GC.
Collapse
Affiliation(s)
- Siyuan Song
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Fang Wen
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Suping Gu
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peixin Gu
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenjie Huang
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Shuai Ruan
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Xiaoxue Chen
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Jiayu Zhou
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Ye Li
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Jiatong Liu
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Shu
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
- *Correspondence: Peng Shu,
| |
Collapse
|