1
|
Jiang L, Yi R, Chen H, Wu S. Quercetin alleviates metabolic-associated fatty liver disease by tuning hepatic lipid metabolism, oxidative stress and inflammation. Anim Biotechnol 2025; 36:2442351. [PMID: 39718035 DOI: 10.1080/10495398.2024.2442351] [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: 08/22/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
The natural flavonoid quercetin, which exhibits a range of biological activities, has been implicated in liver disease resistance in recent research. In vivo study attesting to quercetin's protective effect against metabolic-associated fatty liver disease (MAFLD) is inadequate, however. Here, our investigation explored the potential benefits of quercetin in preventing MAFLD in C57BL/6 mice fed a high-fat diet (HFD). The results revealed that quercetin ameliorated the aberrant enhancement of body and liver weight. The hepatic histological anomalie induced by MAFLD were also mitigated by quercetin. HFD-induced imbalance in serum LDL, HDL, AST, ALT, TG, and LDH was mitigated by quercetin. Mechanically, we found that quercetin improved lipid metabolism by reducing lipogenesis proteins including ACC, FASN, and SREBP-1c and enhancing β-oxidation proteins including PPARα and CPT1A. In vitro study demonstrated that quercetin regulated hepatic lipid metabolism by targeting SREBP-1c and PPARα. Additionally, quercetin enhanced the antioxidant capacity in HFD-treated mice by downregulating Nrf2 and HO-1 expressions and upregulating SOD and GPX1 expressions. The hyper-activation of inflammation was also restored by quercetin via eliminating the phosphorylation of IκBα and NF-κB p65. Collectively, our observations highlight that quercetin exerts hepatoprotective properties in MAFLD mice by regulating hepatic lipid metabolism, oxidative stress and inflammatory response.
Collapse
Affiliation(s)
- Ling Jiang
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Rong Yi
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Huan Chen
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Shuwu Wu
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| |
Collapse
|
2
|
Chen S, Li P, Shi K, Tang S, Zhang W, Peng C, Li T, Xie H, Liu C, Zhou J. Tanshinone IIA promotes ferroptosis in cutaneous melanoma via STAT1-mediated upregulation of PTGS2 expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156702. [PMID: 40222167 DOI: 10.1016/j.phymed.2025.156702] [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: 12/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Melanoma is highly aggressive, metastatic with a poor prognosis. Despite significant advances in targeted therapies and immunotherapies, their efficiency limited by drug resistance. Tanshinone IIA (Tan IIA), a bioactive compound derived from Traditional Chinese plant, exhibits significant anticancer potential, which still needs more research in its complex regulatory mechanisms. PURPOSE This study aimed to elucidate the putative targets and regulatory mechanisms of Tan IIA in anti-melanoma, with a focus on its role in inducing ferroptosis. STUDY DESIGN We designed the experiment to explore the effects of Tan IIA on melanoma through both in vitro and in vivo experiments and to investigate the underlying mechanisms through transcriptomics combining network pharmacology analysis. METHOD Ferroptosis monitored by Malondialdehyde (MDA), Fe2+, reactive oxygen species (ROS) and glutathione (GSH) in vivo and in vitro. RNA sequence was performed to explore the key regulatory pathways involved in Tan IIA-induced ferroptosis. Chromatin immunoprecipitation (ChIP) and Luciferase assays were used to validate transcription factor responsible for prostaglandin-endoperoxide synthase 2 (PTGS2) regulation. Additionally, RT-qPCR, western blot, IF, IHC were aimed to evaluate the expression of target gene. RESULT Tan IIA markedly suppresses melanoma growth in a xenograft model. The same effect performed on inhibition melanoma cells and promotion to ferroptosis with accumulation of ROS, MDA, and Fe²⁺levels and GSH consumption. RNA sequencing and public database analysis revealed that Tan IIA regulates PTGS2, the critical marker of ferroptosis, and PTGS2-knockdown attenuates Tan IIA -induced ferroptosis in melanoma cells. Furthermore, we identified that Tan IIA stimulate signal transducer and activator of transcription 1 (STAT1), a transcription factor, promoting PTGS2 expression and localized in the cell cytoplasm. Moreover, downregulation of the transcription factor STAT1 lead to PTGS2 downregulation and also inhibit ferroptosis in melanoma. CONCLUSION This study, the first to link Tan IIA-induced ferroptosis to the STAT1/PTGS2 axis in melanoma, identifies STAT1 and PTGS2 as novel therapeutic targets for melanoma, which demonstrates the potential of natural compounds Tan IIA in overcoming drug resistance and integrates traditional medicine with advanced molecular techniques for mechanistic exploration.
Collapse
Affiliation(s)
- Shuyue Chen
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Peiting Li
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Ke Shi
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Shijie Tang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wancong Zhang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Tianyu Li
- Department of Burns and Plastic Surgery, Nanshi Hospital of Nanyang, Nanyang, Henan Province 473000, China
| | - Huiqing Xie
- Department of Rehabilitation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Can Liu
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| | - Jianda Zhou
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
3
|
Qiu H, Liu F, Qiu M, Yang J, Peng X. Monotropein attenuates renal cell carcinoma cell progression and M2 macrophage polarization by weakening NF-κB. Int Urol Nephrol 2025; 57:1785-1795. [PMID: 39776402 DOI: 10.1007/s11255-024-04358-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE The study aimed to investigate the effect and mechanism of monotropein on renal cell carcinoma (RCC). METHODS After monotropein and NF-κB receptor activator (RANKL) treatment, cell proliferation, invasion, and apoptosis were evaluated using CCK-8, Transwell, and flow cytometry. Primary macrophages co-cultured with monotropein-treated RCC cells were analyzed to evaluate macrophage polarization using qRT-PCR, western blot, and ELISA assays by detecting the expression of M2 markers (CD206, CD168) and cytokines (IL-10, TGF-β). Additionally, the therapeutic efficacy of monotropein was examined using an RCC mouse xenograft model. RESULTS Monotropein could inhibit the proliferation, invasion, and M2 macrophage polarization and accelerate the apoptosis of RCC cells. Mechanistically, monotropein suppressed NF-κB pathway activation in RCC cells and reduced the expression of NF-κB downstream targets, including Bcl-2, c-Myc, and MMP9. RANKL could eliminate the effect of monotropein on RCC progression. In primary macrophages co-cultured with monotropein-treated RCC cells, monotropein downregulated M2 polarization markers and cytokines, further supporting its role in modulating the tumor microenvironment. In mouse models, monotropein reduced RCC tumor growth, induced apoptosis, and blocked NF-κB pathway. CONCLUSIONS Monotropein prevents RCC malignant progression and reduces M2 macrophage polarization by suppressing the NF-κB pathway, suggesting that monotropein may serve as a potential therapeutic agent for RCC by targeting both tumor cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Heping Qiu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Fei Liu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Mei Qiu
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, 341099, China
| | - Juliang Yang
- Department of Urology, The Affiliated Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou, 341099, China
| | - Xiang Peng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
4
|
Liu WM, Li XB. East meets West: The winning combination against BRAF V600E metastatic colorectal cancer. World J Clin Oncol 2025; 16:102223. [DOI: 10.5306/wjco.v16.i5.102223] [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: 10/12/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 05/19/2025] Open
Abstract
Metastatic colorectal cancer (mCRC) patients with BRAF V600E mutation have a poor prognosis despite the implementation of multiple treatment strategies. The integration of traditional Chinese medicine with Western medicine in treating BRAF mutant mCRC has garnered increasing attention. Recent studies indicate that combining traditional Chinese and modern Western medical approaches not only extend survival but also reduces the risk of mortality in patients with BRAF V600E mutant mCRC. This approach is particularly effective for colorectal cancer patients who have right-sided colon involvement, liver metastasis, or a history of radiotherapy or chemotherapy. In this treatment combination, traditional Chinese medicine may offer symptomatic relief and improve quality of life, while Western medicine targets the disease more aggressively with advanced pharmacological agents. Ongoing research is crucial to further elucidate the mechanisms underlying these benefits and to optimize treatment protocols.
Collapse
Affiliation(s)
- Wen-Ming Liu
- Department of Gastrointestinal Surgery, The First People’s Hospital of Tianmen, Tianmen 431700, Hubei Province, China
| | - Xiao-Bing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, Hubei Province, China
| |
Collapse
|
5
|
Faruk S, Ibrahim KG, Abbas AY, Sulaiman I, Imam MU. Rapamycin and Post-Deficiency Dietary Recovery Reshape Antioxidant Response and Survival in Offspring of Iron-Deficient Mothers. Biol Trace Elem Res 2025:10.1007/s12011-025-04646-6. [PMID: 40338453 DOI: 10.1007/s12011-025-04646-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
Maternal iron deficiency (ID) disrupts maternal and offspring health by impairing iron status and antioxidant defenses. Rapamycin is known to promote autophagy, enhance antioxidant activity, and extend lifespan. This study investigates the intergenerational effects of post-deficiency dietary interventions using normal and rapamycin-treated diets on Drosophila melanogaster. Female flies (F0) were subjected to an iron-deficient diet for 14 days, followed by a 30-day recovery period on either a normal diet or a rapamycin-supplemented diet. Some F0 females were subsequently mated with normal males to produce F1 offspring. Physiological, biochemical, and gene expression analyses were conducted on F0 flies post-chelation and post-intervention. Post-eclosion evaluations, including a 60-day survival study, were performed on both generations. In F0 females, iron chelation significantly reduced (p < 0.0001) body weight, iron levels, and antioxidant enzyme activity, while increasing glutathione (GSH) levels. Gene expression analysis revealed significant changes (p < 0.05) in iron storage (Fer1HCH), autophagy (ATG1), and telomere-related genes (dHeT-A, dTahre, dTart). While a normal diet partially restored iron levels and survival, the rapamycin-treated diet improved antioxidant defenses but had mixed effects on survival and gene expression. In the F1 generation, male and female offspring from mothers on a normal diet exhibited reduced and increased iron levels, respectively, alongside improved median survival. Rapamycin increased body weight and iron levels in female offspring but reduced their median survival. Post-deficiency dietary interventions significantly shape antioxidant responses and survival in both iron-deficient mothers and their offspring. While normal diets support recovery of iron status, rapamycin enhances antioxidant defenses but compromises survival, particularly in female offspring.
Collapse
Affiliation(s)
- Saudatu Faruk
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, 840232, Nigeria
- Department of Biochemistry and Molecular Biology, Faculty of Life and Chemical Sciences, Usmanu Danfodiyo University, P.M.B. 2346, Sokoto, Nigeria
| | - Kasimu Ghandi Ibrahim
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, P.O.Box 2000, Zarqa, 13110, Jordan
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown , Johannesburg, 2193, South Africa
| | - Abdullahi Yahya Abbas
- Department of Biochemistry and Molecular Biology, Faculty of Life and Chemical Sciences, Usmanu Danfodiyo University, P.M.B. 2346, Sokoto, Nigeria
| | - Ismail Sulaiman
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, 840232, Nigeria
- Department of Biochemistry, Faculty of Life Sciences, Kebbi State University of Science and Technology, Aliero, PMB 1144, Kebbi State, Nigeria
| | - Mustapha Umar Imam
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, 840232, Nigeria.
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, 840232, Nigeria.
| |
Collapse
|
6
|
Fang X, Li J, Pang H, Zheng H, Shi X, Feng L, Hu K, Zhou T. Xingxiao pills suppresses lung adenocarcinoma progression by modulating lipid metabolism and inhibiting the PLA2G4A-GLI1-SOX2 Axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156826. [PMID: 40339555 DOI: 10.1016/j.phymed.2025.156826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/20/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) remains a leading cause of cancer mortality due to resistance, metastasis, and recurrence. Unlike conventional cytotoxic therapies, Xingxiao Pills (XXP), a classic traditional Chinese medicine formula, offers a complementary approach to treating LUAD, while its non-cytotoxic anti-cancer mechanisms remain unclear. PURPOSE To investigate the effect and mechanism of XXP on LUAD progression and stemness via lipid metabolism regulation. METHOD UHPLC-MS/MS was used to analyze the chemical constituents of XXP. The effects of XXP on LUAD cell proliferation, migration, invasion, and stemness were evaluated using CCK-8, Transwell, and tumor sphere assays. A LUAD xenograft model confirmed XXP's anti-tumor effects. Transcriptomics, metabolomics, ELISA, qRT-PCR, and Western blot were used to investigate the underlying mechanisms. Kaplan-Meier (KM) survival analysis and stemness index scores were performed for LUAD patients based on the TCGA dataset. Statistical analyses were performed using Student's t-test, ANOVA, and KM survival analysis (p< 0.05 considered significant). RESULTS XXP inhibits LUAD progression in mouse and cell models by targeting lipid metabolism reprogramming. It suppresses FA synthesis, elongation, oxidation, and glycerophospholipid (GPL) metabolism while upregulating arachidonic acid (AA) metabolism. Mechanistic studies revealed that XXP attenuates tumor stemness by inhibiting PLA2G4A (cPLA2), lowering AA release, and disrupting SMO/GLI1/SOX2 signaling, an effect also observed with the cPLA2 inhibitor AACOCF3. KM analysis showed that higher PLA2G4A expression correlated with a worse 5-year prognosis in LUAD (p = 0.0047). The low GPL/high AA group (consistent with XXP's metabolic pattern) had better survival (p = 0.0028) and a lower stemness index (p< 0.0001) than the high GPL/low AA unrelated group. CONCLUSION Xingxiao Pill modulates GPL and AA metabolism and downregulates the PLA2G4A (cPLA2)-AA/SMO/GLI1/SOX2 axis. Through this mechanism, XXP effectively inhibits tumor growth and stemness by targeting lipid metabolism.
Collapse
Affiliation(s)
- Xueni Fang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - JingHua Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - HaoYue Pang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Zheng
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiang Shi
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Kaiwen Hu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Tian Zhou
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
7
|
Yu M, Zhang J, Fu J, Li S, Cui X. Guizhi Fuling decoction protects against bone destruction via suppressing exosomal ERK1 in multiple myeloma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156627. [PMID: 40090044 DOI: 10.1016/j.phymed.2025.156627] [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: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Myeloma-related bone disease (MBD) is a common complication of multiple myeloma (MM) that deteriorates patients' quality of life and affects overall survival. Modulating the interaction between myeloma cells and the bone marrow microenvironment may offer therapeutic potential. While certain natural medicines may regulate bone homeostasis by directly targeting osteoclasts or osteoblasts, few studies have explored the effects of intervening in myeloma cells on osteoclasts, particularly through the role of exosomes. PURPOSE To investigate the inhibitory effect of Guizhi Fuling Decoction (GZFL) on bone lesions formation induced by exosomes secreted by myeloma cells and provide evidence to support the clinical application of GZFL in treating MBD. METHODS TRAP staining and Von Kossa staining were used to evaluate the inhibition of GZFL on RANKL-induced osteoclastogenesis in vitro. Micro-CT and bone histomorphometric analyses were performed to identify the protective effect of GZFL on bone destruction in vivo. RNA immunoprecipitation (RIP), RNA-seq, and UHPLC-MS/MS were conducted to investigate the MBD targets of GZFL. A clinical trial was carried out to evaluate the efficacy of GZFL capsules in the treatment of MBD. RESULTS The main bioactive components of GZFL, paeoniflorin, quercitrin and kaempferol, could target ERK1 and downregulate its expression in MM exosomes. In vitro, GZFL treatment inhibited the promoting effect of MM exosomes on osteoclast (OC) formation, bone resorption, and activated ERK1 expression. In vivo, GZFL prolonged survival rate, inhibited the exacerbation of bone lesions caused by MM exosomes and RANKL-induced ERK1 activation in mice model. Clinical data showed that GZFL capsule combined with bortezomib (Bortezomib) and dexamethasone (PD) significantly reduced the numeric rating scale, as well as the expression levels of ERK and RANKL in bone marrow. ERK1 levels exhibited a positive correlation with both the number of bone lesions and RANKL levels. Higher ERK1 expression indicated a worse prognosis. CONCLUSION GZFL inhibited MBD progression by reducing MM-derived exosomal ERK1, thereby suppressing RANKL-induced ERK1 activation and the downstream OC formation. GZFL combined with PD regimen had good clinical efficacy and safety in the treatment of MBD.
Collapse
Affiliation(s)
- Manya Yu
- The First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, 250014, China
| | - Jie Zhang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250014, China
| | - Jiaqi Fu
- The First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, 250014, China
| | - Suzhen Li
- The First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Jinan, 250014, China
| | - Xing Cui
- Department of Oncology and Hematology, the Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250001, China.
| |
Collapse
|
8
|
Xie J, Shi Z, Sun L, Wu Y, Feng J, Wang H, Lai H. Fangchinoline suppresses nasopharyngeal carcinoma progression by inhibiting SQLE to regulate the PI3K/AKT pathway dysregulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156484. [PMID: 40090046 DOI: 10.1016/j.phymed.2025.156484] [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: 08/24/2024] [Revised: 12/30/2024] [Accepted: 02/06/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND AND PURPOSE Squalene epoxidase (SQLE), a key enzyme in cholesterol metabolism, remains underexplored in nasopharyngeal carcinoma (NPC). Additionally, the therapeutic potential of Fangchinoline, an alkaloid with anticancer properties, has yet to be systematically evaluated. This research investigates Fangchinoline's efficacy in NPC treatment and SQLE-related mechanisms. METHODS Drug screening in NPC cell lines C666-1 and 5-8F identified potential candidates. IC50 values were determined using CCK-8 assays, and apoptosis, proliferation, and invasion were assessed via Annexin V/PI staining, EdU staining, and Transwell assays. Cholesterol levels were quantified using a TG kit. RNA sequencing with GO/KEGG analyses identified key pathways. Correlation analysis was performed via cBioPortal and GEPIA2 databases, protein interaction networks via STRING and Cytoscape, and survival analysis via Kaplan-Meier curves. Gene and protein expression were validated with qPCR and Western blot, and an NPC mouse model confirmed in vivo efficacy. RESULTS Fangchinoline inhibited NPC cell proliferation, induced apoptosis, and reduced cholesterol accumulation. RNA sequencing revealed that Fangchinoline downregulated SQLE expression, suppressing the PI3K/AKT pathway. Correlation and protein interaction analyses highlighted SQLE's role in NPC progression, and survival analysis confirmed its clinical relevance. By targeting SQLE and disrupting cholesterol metabolism, Fangchinoline suppressed tumor growth both in vitro and in vivo. CONCLUSION Our study demonstrates that Fangchinoline inhibits NPC growth by targeting SQLE and disrupting the PI3K/AKT pathway, providing new insights into SQLE as a therapeutic target in NPC.
Collapse
Affiliation(s)
- Jieyun Xie
- Dongguan Hospital of Traditional Chinese Medicine (Department of Internal Medicine II), No. 3 Songshan Lake Avenue East Section, Dongcheng Street, Dongguan City, Guangdong Province, China
| | - Zexian Shi
- Guangzhou University of Chinese Medicine, No. 3 Songshan Lake Avenue East Section, Dongcheng Street, Dongguan City, Guangdong Province, China
| | - Lingling Sun
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine (Oncology Department), No. 16 Airport Road, Baiyun District, Guangzhou City, Guangdong Province, China
| | - Yihong Wu
- Dongguan Hospital of Traditional Chinese Medicine (Department of Internal Medicine II), No. 3 Songshan Lake Avenue East Section, Dongcheng Street, Dongguan City, Guangdong Province, China
| | - Jiuhuan Feng
- Dongguan Hospital of Traditional Chinese Medicine (Department of Internal Medicine II), No. 3 Songshan Lake Avenue East Section, Dongcheng Street, Dongguan City, Guangdong Province, China
| | - Han Wang
- Dongguan Hospital of Traditional Chinese Medicine (Department of Internal Medicine II), No. 3 Songshan Lake Avenue East Section, Dongcheng Street, Dongguan City, Guangdong Province, China
| | - Haifeng Lai
- Dongguan Hospital of Traditional Chinese Medicine (Department of Internal Medicine IX), No. 3 Songshan Lake Avenue East Section, Dongcheng Street, Dongguan City, Guangdong Province, China.
| |
Collapse
|
9
|
Liu Y, Yang J, Yu F, Li L, Zhao N, Lu C, Lu A, He X. Research advances in traditional Chinese medicine formulae and active components targeting lipid metabolism for hepatocellular carcinoma therapy. Front Pharmacol 2025; 16:1528671. [PMID: 40351413 PMCID: PMC12062747 DOI: 10.3389/fphar.2025.1528671] [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: 11/15/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) has a relatively poor prognosis and a high degree of malignancy. However, the therapeutic drugs are limited. In recent years, abnormal lipid metabolism and its important role in HCC has been reported, and emerging studies found that some formulae and active components of traditional Chinese medicine (TCM) can regulate abnormal lipid metabolism in HCC, showing their good application prospects. Therefore, this article summarizes the changes and the roles of lipid metabolites in HCC progression, and discusses the role of formulae and active components of TCM for the treatment of HCC based on their regulation on abnormal lipid metabolism. A deeper understanding of their relationship may help the precise use of these formulae and active components in HCC.
Collapse
Affiliation(s)
- Yang Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Yang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fenghua Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Shanghai GuangHua Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Li Q, Wang M, Huang X, Wang S, Li C, Li P, Xiang W, Yao L, Deng C, Zhang M, Wang J. 6-Gingerol, an active compound of ginger, attenuates NASH-HCC progression by reprogramming tumor-associated macrophage via the NOX2/Src/MAPK signaling pathway. BMC Complement Med Ther 2025; 25:154. [PMID: 40269843 PMCID: PMC12020160 DOI: 10.1186/s12906-025-04890-2] [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: 10/27/2024] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Non-alcoholic steatohepatitis-associated hepatocellular carcinoma (NASH-HCC) accounts for an increasing proportion of HCC cases. Currently, effective pharmacological options for treating both NASH and NASH-HCC remain limited, necessitating the identification of novel therapeutic agents. Our previous studies have demonstrated that ginger can ameliorate nonalcoholic fatty liver disease (NAFLD) and prevent the occurrence of NASH. The therapeutic effects and underlying mechanisms of NASH-HCC, however, remain poorly understood. METHODS Network pharmacology, bioinformatics, single-cell RNA sequencing analysis, and molecular docking were used to identify the main active compounds, targets, and possible mechanisms of ginger in treating NASH-HCC. The anti-tumor efficacy and underlying mechanisms of the selected compound in treating NASH-HCC were validated through in vitro experimentation. RESULTS Network pharmacology, bioinformatics, and molecular docking have revealed that 6-gingerol is the main active compound of ginger in treating NASH-HCC. SRC can be an essential target gene for ginger attenuating NASH-HCC progression, while the mitogen-activated protein kinase (MAPK) signaling pathway and reactive oxygen species (ROS) play equally important roles. Single-cell RNA sequencing of the HCC patients shows that the key targets of ginger in treating NASH-HCC are distributed in tumor-associated macrophage (TAMs). It has been reported that NOX2-derived ROS in macrophages can activate Src and then regulate downstream MAPK signaling cascades. 6-Gingerol can inhibit the proliferation, migration and reduce lipid deposition of liver cancer cells in vitro. More importantly, it induces polarization TAMs to M1 and enhances proinflammatory function, which may be achieved via the NOX2/Src/MAPK signaling pathway. CONCLUSION This study proves that 6-gingerol, the primary active compound in ginger, plays a role in attenuating the progression of NASH-HCC by inhibiting the proliferation and migration of tumor cells, or reprogramming TAMs to the M1 phenotype via the NOX2/Src/MAPK signaling pathway and activating the TAM-mediated immune responses.
Collapse
Affiliation(s)
- Qiurui Li
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Meng Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Xuekuan Huang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Shang Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Chunli Li
- School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Pan Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Wei Xiang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China
| | - Ling Yao
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China
| | - Chengdan Deng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China
| | - Mingming Zhang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China.
| | - Jianwei Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing University of Chinese Medicine, No. 61 Puguobao Road, Bishan District, Chongqing, 402760, China.
| |
Collapse
|
11
|
Zheng X, Shi C, Xie Y, Wen Q, Lyu T, Li H, Wang Z, Shen M, Zhu Y, Ruan S. Bioactive components of Jiedu Sangen decoction against colorectal cancer: A novel and comprehensive research strategy for natural drug development. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156795. [PMID: 40279966 DOI: 10.1016/j.phymed.2025.156795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Jiedu Sangen Decoction (JSD) is widely used in the treatment of colorectal cancer (CRC) patients in southern China due to its good clinical efficacy, but the effective active ingredients are still unknown. PURPOSE This study fully explored the bioactive components of JSD based on an innovative and comprehensive research strategy. Using advanced computer technology (e.g., machine learning AHP-SOM algorithm and molecular dynamics simulation) to identify the most promising bioactive components and key targets in JSD, in order to provide new perspectives for the development of natural drugs. METHODS UPLC-MS/MS was used to screen bioactive components in JSD and rat plasma, and network pharmacology analysis combined with machine learning yielded the most promising bioactive components. RNA-seq was used to screen therapeutic targets before and after JSD acted on SW620 cells, and bioinformatics was used to analyze the clinical significance of these key targets. Molecular docking, molecular dynamics simulation, and experiments verified the most promising bioactive components and their therapeutic targets. RESULTS JSD exhibited a strong pro-apoptotic effect on CRC in vitro. UPLC-MS/MS screened out 18 prototype components and 8 possible metabolites of JSD entering the blood. Network pharmacology combined with machine learning identified the three most promising bioactive components. RNA sequencing and bioinformatics analysis revealed six key targets of JSD against CRC. Molecular docking and molecular dynamics simulations proposed the most promising "small molecule drug-target protein" combinations, and SPR and MST demonstrated the direct binding between them: Resveratrol - CA9, Genistein - NOTUM, and Afzelin - DPEP1. Molecular biology experiments found that resveratrol may promote CRC apoptosis through the CA9/PI3K/AKT signaling pathway, and genistein targets NOTUM to downregulate β-catenin expression to inhibit CRC proliferation. CONCLUSION It is feasible to develop a novel and comprehensive research strategy to fully explore bioactive components of JSD and provide full support for natural drug development.
Collapse
Affiliation(s)
- Xueer Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China
| | - Chao Shi
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China
| | - Ying Xie
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Qing Wen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China
| | - Tongdan Lyu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China
| | - Hao Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China
| | - Zhenru Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China
| | - Minhe Shen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China.
| | - Ying Zhu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China.
| | - Shanming Ruan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, PR China.
| |
Collapse
|
12
|
Liu L, Xing G, Guo X, Chen H, Li J, Wang J, Li Y, Liang G, Liu M. Inhibition of colorectal cancer cell growth by downregulation of M2-PK and reduction of aerobic glycolysis by clove active ingredients. Front Pharmacol 2025; 16:1552486. [PMID: 40308769 PMCID: PMC12041220 DOI: 10.3389/fphar.2025.1552486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Exploring the anti-tumor molecular mechanisms of traditional Chinese medicines has become an important strategy to develop novel anti-tumor drugs in the clinic. Several pharmacological studies have reported the antioxidant, antibacterial, anti-inflammatory, and anti-tumor effects of clove. Previously, we have shown that the active fraction from clove (AFC) can inhibit the growth of tumor cells, particularly colon cancer cells, in vitro. However, the mechanism of action regarding the anti-colon cancer activity of AFC, especially in aerobic glycolysis, has not been adequately investigated. In this study, we found that AFC significantly inhibited the growth of five types of colon cancer cells, downregulated the mRNA and protein levels of M2-type pyruvate kinase (PKM2), and reduced aerobic glycolysis capacity. Transfection of PKM2-siRNA mimicked the inhibitory effects of AFC on aerobic glycolysis in colon cancer cells. Furthermore, the highly expressed, tumor-specific targets c-myc and cyclin D1 in cells were also found to be downregulated following the action of AFC. In the HCT116 cell xenograft nude mice models, the results after AFC administration were consistent with those of the cellular experiments, while AFC caused less liver injury and weight loss than the conventional chemotherapeutic agent 5- fluorouracil (5-FU). In conclusion, AFC inhibits colon cancer growth by downregulating PKM2 to inhibit aerobic glycolysis and reduce the tumor-specific high expression of c-myc and cyclin D1. Future work should explore how it downregulates pyruvate kinase (PK) in the first place, along with the intrinsic mechanism between the downregulation of PKM2 and the downregulation of c-myc.
Collapse
Affiliation(s)
- Lin Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Drug Dispending Department, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyi Guo
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Li
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jian Wang
- Discipline Construction Office, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yaling Li
- Pharmacy Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Liang
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
13
|
Guo Q, Mao Y, Zhang J, Zhou Y, Zhao Y, Li Y, Lv J, Yang H, Liu B. Oridonin combined with cisplatin synergistically induces apoptosis by activating the NOXA-BCL2 axis in esophageal squamous cell carcinoma. Biochem Pharmacol 2025; 237:116953. [PMID: 40250733 DOI: 10.1016/j.bcp.2025.116953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/04/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Esophageal cancer, a malignant neoplasm originating from the epithelial cells of the esophagus, predominantly manifests as esophageal squamous cell carcinoma (ESCC) in approximately 90% of cases in China. Cisplatin-based chemotherapy regimens remain the first-line therapeutic option for ESCC, however, the five-year overall survival rate of patients is disappointingly low. Oridonin, a bioactive diterpenoid extracted from the traditional Chinese medicine herb Donglingcao, has demonstrated inhibitory effects against various malignancies. Currently, research on the combination of oridonin and cisplatin for the treatment of ESCC is limited. This study aims to elucidate the potential synergistic anti-cancer effects of oridonin in combination with cisplatin on ESCC, along with the underlying synergistic molecular mechanisms. In vitro experiments revealed that the combination of oridonin and cisplatin could synergistically inhibit ESCC cell proliferation, migration, invasion. The synergistic effect also induced cell cycle arrest and promoted apoptosis via the mitochondrial pathway by augmenting NOXA transcriptional activity and activating the NOXA-BCL2 axis. In vivo experiments corroborated these findings, showing a marked reduction in the growth of subcutaneous xenograft tumors in mice treated with the combination, without exacerbating the cisplatin-associated side effects such as weight loss or hepatic and renal toxicity. In conclusion, the combination of oridonin and cisplatin can synergistically inhibit the development of ESCC through the activation of the NOXA-BCL2 axis signaling pathway. This treatment is both safe and effective,presenting a promising prospect for combined therapeutic application in ESCC management.
Collapse
Affiliation(s)
- Qihang Guo
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052 Henan, China
| | - Yue Mao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052 Henan, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052 Henan, China
| | - Jiyu Zhang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052 Henan, China
| | - Yangyang Zhou
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000 Hubei, China
| | - Yue Zhao
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052 Henan, China
| | - Ying Li
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052 Henan, China
| | - Jinglong Lv
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052 Henan, China
| | - Huiyu Yang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052 Henan, China.
| | - Bingrong Liu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052 Henan, China.
| |
Collapse
|
14
|
Ichikawa K, Johnson HM, Curtis MA, Biswas N, Singh S, Khachatryan HN, Gater AE, Lin SX, Sperry J. Targeting glioma with heteroaromatic alkaloids: A review of potential therapeutics. Bioorg Med Chem 2025; 121:118051. [PMID: 39999647 DOI: 10.1016/j.bmc.2024.118051] [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: 09/20/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 02/27/2025]
Abstract
Glioblastoma multiforme (GBM), classified as a grade IV astrocytoma, is the most aggressive and deadly form of glioma, characterized by rapid progression, extensive genetic heterogeneity, and resistance to conventional therapies. Despite advancements in surgical techniques, radiation therapy, and the frontline chemotherapeutic agent temozolomide, the prognosis for GBM patients remains poor, with a median survival of 15 months and a 5-year survival rate of approximately 7 %. The absence of effective long-term treatments underscores the urgent, unmet clinical need for novel therapeutic strategies to improve patient outcomes. Natural products, particularly alkaloids, have garnered attention as a rich source of bioactive compounds with diverse pharmacological properties. Alkaloids, a structurally diverse group of natural products, are renowned for their chemotherapeutic properties and ability to cross the blood-brain barrier (BBB), making them promising candidates for glioma therapy. This review systematically examines all reported heteroaromatic alkaloids with documented anti-glioma activities, highlighting their mechanisms of action where available. By providing a comprehensive resource, it aims to facilitate the identification and optimisation of alkaloid-based compounds for glioma-targeted drug discovery. Additionally, this review emphasizes the importance of incorporating natural products into the drug development pipeline to address the pressing challenges associated with glioma, particularly GBM treatment.
Collapse
Affiliation(s)
- Karen Ichikawa
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand; Centre for Brain Research, University of Auckland, Auckland, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Hannah M Johnson
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, University of Auckland, Auckland, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Nandita Biswas
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Hasmik N Khachatryan
- Scientific Technological Centre of Organic and Pharmaceutical Chemistry, National Academy of Science of Armenia, Yerevan 0014, Armenia
| | - Anastasia E Gater
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Simon X Lin
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Jonathan Sperry
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand.
| |
Collapse
|
15
|
Zhu Z, Lian X, Hu J, Wang Z, Zhong Y, Zhao Y, Lu L, Pan Y, Zhou M, Xu J. DPHC from Alpinia officinarum Hance specifically modulates the function of CENPU in the cell cycle and apoptosis to ameliorate hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119598. [PMID: 40058474 DOI: 10.1016/j.jep.2025.119598] [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: 01/20/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alpinia officinarum Hance (A. officinarum), a perennial herb used in the treatment of digestive system cancers, holds significant value for the Li people of Hainan as a traditional Chinese medicine. (R)-5-hydroxy-1,7-diphenyl-3-heptanone (DPHC), a diarylheptanoid component is derived from A. officinarum. Diarylheptanoids have demonstrated anti-proliferative effects on breast cancer cells, neuroblastoma cells, and other tumor cells. However, the pharmacological activity of DPHC in improving hepatocellular carcinoma (HCC) remains undefined. AIM OF THE STUDY To elucidate the anti-HCC effects of DPHC derived from A. officinarum and explore its underlying mechanistic pathways both in vivo and in vitro. MATERIAL AND METHODS The effects of DPHC on HCC cell lines were evaluated in vitro using cell counting kit-8, EdU cell proliferation assays, a wound healing assay, a three-dimensional tumor spheroid model, and flow cytometry. The ability of DPHC to ameliorate HCC was assessed in vivo via a nude mouse subcutaneous xenograft tumor model, serum biochemical marker detection, and hematoxylin-eosin staining. The molecular mechanism of DPHC in HCC was elucidated through a combination of transcriptome sequencing, cell transfection, immunohistochemistry assay, immunofluorescence staining, quantitative reverse transcription-PCR, and western blot analysis. RESULTS DPHC induced significant G0/G1 phase arrest and apoptosis in HepG2 and HCCLM3 cells while also markedly inhibiting tumor growth in nude mice. Mechanically, DPHC directly interacted with centromere-associated protein U (CENPU) to suppress its expression. The reduced expression of CENPU results in decreased interaction with the transcription factor E2F6, thereby affecting the transcriptional activity of the transcription factor E2F1. This subsequently inhibits the expression of downstream cell cycle factors (CCND1, CDK4, and CDK1) and increases apoptosis factors (Caspase 3 and Caspase 9). CONCLUSIONS DPHC from A. officinarum specifically modulates the function of CENPU in the cell cycle and apoptosis to ameliorate HCC. Our study revealed the anti-HCC effect and underlying mechanism of DPHC, offering new insights and potential targets for HCC treatment.
Collapse
Affiliation(s)
- Zhe Zhu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Xiuxia Lian
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education & International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province & Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, Hainan, 571199, China
| | - Jicheng Hu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Zhe Wang
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yinghong Zhong
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yuan Zhao
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Lu Lu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yipeng Pan
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| | - Mingyan Zhou
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| | - Jian Xu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China; Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
16
|
Jin W, Zhang Y, Pang S, Yao D, Huang Y. Off-label use of sodium cantharidinate and vitamin B6 injection in cancer: a protocol for a systematic review and meta-analysis. Syst Rev 2025; 14:82. [PMID: 40205599 PMCID: PMC11984136 DOI: 10.1186/s13643-025-02826-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND In China, sodium cantharidinate/vitamin B6 (SC/VB6) injection has been approved since 2002 for the treatment of lung cancer and primary liver cancer. In addition to these authorized indications, clinical application of SC/VB6 is also common in various other types of cancer. However, there is a lack of comprehensive understanding on this topic. Thus, this systematic review and meta-analysis aims to consolidate evidence regarding the efficacy and safety of off-label use of SC/VB6 in oncology. METHODS International databases including PubMed, Embase, Cochrane Library, Web of Science, and CINAHL Plus, as well as Chinese databases including China National Knowledge Infrastructure (CNKI), Chinese Biomedical Literature Database (CBM), and Wanfang, will be searched from the inception to 31 December 2024. Comparative studies that evaluated the add-on effect of SC/VB6 to conventional cancer treatments against the use of conventional treatments alone will be considered in the scope of this review. The primary outcomes are objective response rate and performance status. Secondary outcomes are disease control rate (DCR), progression-free survival (PFS), disease-free survival (DFS), overall survival (OS), and adverse events (AEs). Depending on heterogeneity, data will be synthesized using either the Mantel-Haenszel fixed-effect or the DerSimonian and Laird random-effect model. Subgroup analyses will be conducted for the following variables: type of cancer, study design, SC/VB6 dosage, treatment duration, and combined therapies, provided that each subgroup contains at least two studies. Sensitivity analyses will be performed on efficacy outcomes. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) will be utilized to appraise the overall quality of evidence. DISCUSSION This review will encompass both randomized controlled trials (RCTs) and cohort studies, thereby enabling us to synthesize and assess evidence across experimental and real-world observational settings. Our findings will contribute to a better understanding on the benefit-risk profile regarding the off-label use of SC/VB6 in oncology, guiding the trajectory of future research, and offering a robust scientific foundation to inform clinical and regulatory decision-making process. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42024504977.
Collapse
Affiliation(s)
- Wenzhen Jin
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China
| | - Yibin Zhang
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China
| | - Shu Pang
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China
| | - Dongdong Yao
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China
| | - Yiwen Huang
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China.
| |
Collapse
|
17
|
Han H, Wen Z, Yang M, Wang C, Ma Y, Chen Q, Jiang D, Xu Y, Fazal A, Jie W, Lv X, Yin T, Lin H, Lu G, Qi J, Yang Y, Xu G. Shikonin Derivative Suppresses Colorectal Cancer Cells Growth via Reactive Oxygen Species-Mediated Mitochondrial Apoptosis and PI3K/AKT Pathway. Chem Biodivers 2025; 22:e202403291. [PMID: 40022742 DOI: 10.1002/cbdv.202403291] [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: 12/11/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025]
Abstract
Colorectal cancer (CRC) is one of the deadliest cancers globally, ranking as the third most prevalent and second most lethal malignancy worldwide. The standard treatment for CRC typically involves a combination of surgery, radiotherapy, and chemotherapy. Despite advancements in CRC treatment, the prognosis remains unsatisfactory, primarily due to unclear mechanisms underlying tumorigenesis and the aggression of CRC. The aberrant activation of the PI3K/AKT pathway is frequently implicated in the initiation, progression, and metastasis of CRC. Studies have demonstrated that shikonin (SK) exerts anti-cancer effects. In this study, we evaluated the anti-tumor activities of a series of semi-synthesized SK derivatives against CRC cells. Our findings revealed that the SK derivative (M12) significantly inhibited the proliferation and colony formation of CRC cells, reduced cell migration, and induced apoptosis. Mechanistically, M12 enhanced the production of reactive oxygen species and downregulated the mitochondrial membrane potential, ultimately leading to mitochondrial apoptosis. Furthermore, M12 exhibited anti-CRC effects by modulating the PI3K/AKT signaling pathway and significantly suppressed tumorigenicity without causing notable adverse effects in mice. Therefore, targeting the PI3K/AKT pathway could be a promising treatment for CRC. M12 appears to be a promising candidate for the effective and safe treatment of CRC.
Collapse
Affiliation(s)
- Hongwei Han
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Zhongling Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Minkai Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Changyi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Yudi Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Qingqing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Dexing Jiang
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
| | - Ye Xu
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
| | - Aliya Fazal
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Wencai Jie
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Xiaoran Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Tongming Yin
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Hongyan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Guihua Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, China
| | - Jinliang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Yonghua Yang
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, China
| | - Guohua Xu
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
18
|
Saikia L, Talukdar NC, Dutta PP. Exploring the Therapeutic Role of Flavonoids Through AMPK Activation in Metabolic Syndrome: A Narrative Review. Phytother Res 2025; 39:1403-1421. [PMID: 39789806 DOI: 10.1002/ptr.8428] [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: 05/19/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 01/12/2025]
Abstract
Metabolic syndrome (MetS) is a cluster of interrelated metabolic abnormalities that significantly elevate the risk of cardiovascular disease, obesity, and diabetes. Flavonoids, a diverse class of bioactive polyphenolic compounds found in plant-derived foods and beverages, have garnered increasing attention as potential therapeutic agents for improving metabolic health. This review provides a comprehensive analysis of the therapeutic effects of flavonoids in the context of the MetS, with a particular focus on their modulation of the AMP-activated protein kinase (AMPK) pathway. AMPK serves as a central regulator of cellular energy balance, glucose metabolism, and lipid homeostasis, making it a critical target for metabolic intervention. Through a systematic review of the literature up to April 2024, preclinical studies across various flavonoid subclasses, including flavonols, and flavan-3-ols, were analysed to elucidate their mechanistic roles in metabolic regulation. Many studies suggests that flavonoids enhance glycolipid metabolism by facilitating glucose transporter 4 (GLUT4) translocation and activating the AMPK pathway, thereby improving glycemic control in diabetes models. In obesity-related studies, flavonoids demonstrated significant inhibitory effects on lipid synthesis, reduced adipogenesis, and attenuated proinflammatory cytokine secretion via AMPK activation. These findings show the broad therapeutic potential of flavonoids in addressing the MetS and its associated disorders. While these preclinical insights highlight flavonoids as promising natural agents for metabolic health improvement, it is important to note that their excessive concentrations may disrupt these pathways, potentially leading to metabolic imbalance and cytotoxicity. Further studies and clinical trials are essential to determine optimal dosing regimens, formulations, and the long-term safety and efficacy of flavonoids. This review highlights the importance of flavonoids for natural interventions targeting MetS and its comorbidities, offering a foundation for future translational research.
Collapse
Affiliation(s)
- Lunasmrita Saikia
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, India
| | | | - Partha Pratim Dutta
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, India
| |
Collapse
|
19
|
Liu Z, Wang Y, Gao X, Ma J, Hui C, Wang C, Liu Y, Huang Y, Wen Y. Tanshinone IIA Suppresses the Proliferation of MGC803 Cells by Disrupting Glycolysis Under Anaerobic Conditions. Appl Biochem Biotechnol 2025:10.1007/s12010-025-05205-4. [PMID: 40009338 DOI: 10.1007/s12010-025-05205-4] [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] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
This study aimed to investigate how Tanshinone IIA (Tan IIA) affects gastric cancer cell (MGC803) proliferation under anaerobic conditions, which are linked to drug resistance and tumor growth. The proliferation of MGC803 cells under both aerobic and anaerobic conditions in response to Tan IIA was assessed using the Cell Counting Kit-8 (CCK-8) assay. To elucidate the molecular mechanisms underlying these effects, proteomics analysis was performed following treatment with 50 µmol/L Tan IIA, focusing on alterations in Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Additionally, in vitro evaluations such as glucose uptake, lactate production, and adenosine triphosphate (ATP) synthesis were employed to validate the alterations in glycolytic activity observed in anaerobic cells treated with Tan IIA. Under anaerobic conditions, Tan IIA enhanced the inhibitory effect on the proliferation of MGC803 cells. Proteomics data revealed that a total of 6629 proteins were identified and quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS), with 2604 proteins exhibiting significant changes (fold change > 2 or < 0.5, P < 0.05). KEGG analysis highlighted the perturbation of glycolytic pathway by Tan IIA under anaerobic conditions, accompanied by reduced glucose uptake, lactate production, and ATP synthesis. Additionally, a downregulation of glycolytic enzyme expression was observed at both the mRNA and protein levels, including glyceraldehyde-3-phosphate dehydrogenase (GAPDH), lactate dehydrogenase A (LDHA), phosphofructokinase 2 (PFKP), and pyruvate dehydrogenase (PDH). Tan IIA inhibits the proliferation of MGC803 cells by disrupting the glycolysis under anaerobic conditions, offering a potential treatment for anaerobiosis-resistant solid tumors.
Collapse
Affiliation(s)
- Zhe Liu
- Department of Pathology, The Ninth Hospital of Xi'an, 710054, Xi'an, Shaanxi, People's Republic of China
| | - Yi Wang
- Department of Pathology, The Ninth Hospital of Xi'an, 710054, Xi'an, Shaanxi, People's Republic of China
| | - Xia Gao
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, 710038, Xi'an, Shaanxi, People's Republic of China
| | - Jingwen Ma
- Radiology Department, CT and MRI Room, The Ninth Hospital of Xi'an, 710054, Xi'an, Shaanxi, People's Republic of China
| | - Chan Hui
- Department of Pathology, The Ninth Hospital of Xi'an, 710054, Xi'an, Shaanxi, People's Republic of China
| | - Chao Wang
- Military Hospital, Unit 94162 of the Chinese people's Liberation Army, 710600, Xi'an, Shaanxi, People's Republic of China
| | - Yanfei Liu
- Department of Pathology, The Affiliated Children's Hospital of Xi'an Jiaotong University, 710003, Xi'an, Shaanxi, People's Republic of China
| | - Yao Huang
- Department of Oncology, The Ninth Hospital of Xi'an, 710054, Xi'an, Shaanxi, People's Republic of China.
| | - Yuting Wen
- Department of Pathology, The Ninth Hospital of Xi'an, 710054, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
20
|
Guo TH, Hong SW, Zhu WJ, Hui YF, Qiu WL, Wu Y, Li X, Ke F, Li L, Cheng HB. Anti-programmed death-1 immunotherapy-promising treatment for metastatic colorectal cancer: A case report. World J Gastrointest Oncol 2025; 17:100954. [PMID: 39958538 PMCID: PMC11756018 DOI: 10.4251/wjgo.v17.i2.100954] [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: 08/31/2024] [Revised: 10/30/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most prevalent form of cancer worldwide. Among patients with CRC, colorectal liver metastasis (CRLM) is the foremost direct contributor to mortality. In recent years, immunotherapy has swiftly risen to prominence as a vital approach for treating a range of solid tumors, including CRC. We present a unique case of a patient suffering from CRLM, with the goal of offering an insightful example and relevant references for the treatment of CRLM. CASE SUMMARY We report a patient who experienced liver metastasis after undergoing successful surgical removal of CRC, with the postoperative pathological stage identified as pT4N2aM0. The patient has been receiving a combination treatment of Western and Traditional Chinese Medicine. Regular assessments of the patient's condition have been conducted, encompassing evaluations of serum carcinoembryonic antigen levels, carbohydrate antigen 199, and observations of the tongue complexion and its coating. The patient achieved clinical remission after anti-programmed death-1 immunotherapy when various systemic therapies failed. Since the diagnosis of CRLM, the patient has survived for more than 6 years, surpassing the expected survival time for those with advanced CRC. CONCLUSION This case illustrates the considerable promise of anti-programmed death-1 immunotherapy in managing CRLM, especially in scenarios of drug resistance and disease progression.
Collapse
Affiliation(s)
- Tian-Hao Guo
- Institute of Health and Regimen, Jiangsu Open University, Nanjing 210036, Jiangsu Province, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Sheng-Wei Hong
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Wen-Jian Zhu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Yi-Fan Hui
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Wen-Li Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Yan Wu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Xuan Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Fei Ke
- Department of Pathology, Jiangsu Provincial Hospital of Traditional Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing 210029, Jiangsu Province, China
| | - Liu Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Hai-Bo Cheng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
21
|
Carrasco M, Guzman L, Olloquequi J, Cano A, Fortuna A, Vazquez-Carrera M, Verdaguer E, Auladell C, Ettcheto M, Camins A. Licochalcone A prevents cognitive decline in a lipopolysaccharide-induced neuroinflammation mice model. Mol Med 2025; 31:54. [PMID: 39930360 PMCID: PMC11812219 DOI: 10.1186/s10020-025-01106-8] [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: 07/31/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
Inflammation plays a key role in the development of neurodegenerative disorders that are currently incurable. Licochalcone A (LCA) has been described as an emerging anti-inflammatory drug with multiple therapeutical properties that could potentially prevent neurodegeneration. However, its neuroprotective mechanism remains unclear. Here, we investigated if LCA prevents cognitive decline induced by Lipopolysaccharide (LPS) and elucidated its potential benefits. For that, 8-week-old C57BL6/J male mice were intraperitonially (i.p.) treated with saline solution or LCA (15 mg/kg/day, 3 times per week) for two weeks. The last day, a single i.p injection of LPS (1 mg/kg) or saline solution was administered 24 h before sacrifice. The results revealed a significant reduction in mRNA expression in genes involved in oxidative stress (Sod1, Cat, Pkm, Pdha1, Ndyfv1, Uqcrb1, Cycs and Cox4i1), metabolism (Slc2a1, Slc2a2, Prkaa1 and Gsk3b) and synapsis (Bdnf, Nrxn3 and Nlgn2) in LPS group compared to saline. These findings were linked to memory impairment and depressive-like behavior observed in this group. Interestingly, LCA protected against LPS alterations through its anti-inflammatory effect, reducing gliosis and regulating M1/M2 markers. Moreover, LCA-treated animals showed a significant improvement of antioxidant mechanisms, such as citrate synthase activity and SOD2. Additionally, LCA demonstrated protection against metabolic disturbances, downregulating GLUT4 and P-AKT, and enhanced the expression of synaptic-related proteins (P-CREB, BDNF, PSD95, DBN1 and NLG3), leading all together to dendritic spine preservation. In conclusion, our results demonstrate that LCA treatment prevents LPS-induced cognitive decline by reducing inflammation, enhancing the antioxidant response, protecting against metabolic disruptions and improving synapsis related mechanisms.
Collapse
Affiliation(s)
- Marina Carrasco
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Laura Guzman
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Jordi Olloquequi
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Amanda Cano
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ana Fortuna
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT/ICNAS, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Manuel Vazquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Networking Research Centre of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28031, Madrid, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), University of Barcelona, 08028, Barcelona, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain.
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| |
Collapse
|
22
|
Rui M, Su Y, Tang H, Li Y, Fang N, Ge Y, Feng Q, Feng C. Computational Design and Optimization of Multi-Compound Multivesicular Liposomes for Co-Delivery of Traditional Chinese Medicine Compounds. AAPS PharmSciTech 2025; 26:61. [PMID: 39934607 DOI: 10.1208/s12249-025-03042-6] [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: 08/19/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
Study explored the synergistic anti-tumor effects of a combination of compounds from Traditional Chinese Medicine, including rosmarinic acid (RA), chlorogenic acid (CA), and scoparone (SCO), in the formulation of multivesicular liposomes (MVLs). Optimization of formulations and process parameters was essential to achieve effective liposomal encapsulation and optimal release profiles for these three compounds with diverse properties. Traditional trial-and-error approaches are inefficient for the optimization of complex multi-compound MVLs. We developed a new formulation optimization model, which could address this issue by predicting the optimal multi-compound MVLs formulation. Our machine learning model integrated support vector machine regression (SVR) algorithm and cuckoo search (CS) algorithm, resulting in three CS-SVR models to predict single-compound MVLs. The CS algorithm, with various weighting rules, was then applied to search the best formulation parameters across three CS-SVR models and to maximize the encapsulation efficiency for all three compounds. The multi-compound MLVs were subsequently prepared under the predicted conditions, achieving an optimized particle size of 15.12 µm, with encapsulation efficiencies of 82.93 ± 2.43% for CA, 82.22 ± 1.25% for RA, and 95.60 ± 0.18% for SCO. The predicted optimal multi-compound MVLs were further validated through in vitro characterization and in vivo anti-tumor experiments, showing a promising synergistic anti-tumor effect consistent with in vitro results. This model accurately predicted optimal encapsulation conditions, which were validated experimentally, demonstrating improved encapsulation efficiencies and reduced trial-and-error iterations. Collectively, our model provides a predictive pathway for multi-compound MVLs formulation, indicating the ability of this model to significantly reduce experimental burden and accelerate formulation development.
Collapse
Affiliation(s)
- Mengjie Rui
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Yali Su
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Haidan Tang
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Yinfeng Li
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Naying Fang
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Yingying Ge
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Qiuqi Feng
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Chunlai Feng
- Department of Obstetrics, Affiliated Hospital of Jiangsu University, No.438 Jiefang Road, Zhenjiang, 212001, Jiangsu Province, China.
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China.
| |
Collapse
|
23
|
Hou X, Liang X, Zhao X, Shi Y, Zhuo F, Tong X, Yang X, Zhai Q, Wang J, Guo Q, Tu P, Zeng K, Zhang Q. Uncaria rhynchophylla alkaloid extract exerts neuroprotective activity against Parkinson's disease via activating mitophagy with the involvement of UCHL1. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119009. [PMID: 39471877 DOI: 10.1016/j.jep.2024.119009] [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: 05/07/2024] [Revised: 09/25/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Uncaria rhynchophylla (Miq.) Miq. ex Havil. (UR), a traditional Chinese medicinal plant, plays an active role in neuroprotection. Clinical medication and modern pharmacological studies have proved the efficacy of UR against Parkinson's disease (PD), with alkaloids being recognized as the main bioactive components. But the therapeutic effect and mechanism of U. rhynchophylla alkaloid extract (URA) against PD need further exploration. AIM OF STUDY The study aimed to investigate the therapeutic effect and potential mechanism of URA on PD. MATERIALS AND METHODS LC-MS methodology was used to evaluate the chemical constituents of URA. The anti-PD activity of URA in vivo was measured on the mouse and rat models of PD. Neuroprotective effect of URA on PC12 cells was measured by MTT assay. Dopamine (DA) and its metabolites were detected by LC-MS for probing the protection ability on dopaminergic neurons. The differentially expressed proteins between model group and URA administrated group were analyzed by proteomics, suggesting oxidative phosphorylation as possible pathway of URA. Considering the critical role of mitochondria in oxidative phosphorylation, JC-1 staining, MitoSOX staining, transmission electron microscopy (TEM) observation and adenosine triphosphate (ATP) levels detection were used to analyze the effects of URA on mitochondrial morphology and function. Biolayer interferometry (BLI) was used to search for the possible UCHL1-bonding compounds in URA. RESULTS URA significantly mitigated the behavioural defects by improving coordination, shortening the time to climb down the whole pole (T-LA) and increasing the forelimbs' muscle strength of MPTP-induced PD mice and 6-OHDA-induced PD rats. In addition, URA improved tyrosine hydroxylase expression in dopaminergic neurons by immunohistochemistry (IHC) staining, and thus increased the neurotransmitter levels of DA and relevant metabolites. Furthermore, URA promoted mitophagy as reflected by a significant decrease in reactive oxygen species (ROS) generation, an increase in ATP levels and clearance of damaged mitochondria. Subsequently, Ubiquitin C-terminal hydrolase 1 (UCHL1), which is associated with the mitochondrial dysfunction in PD, is suggested to be a promising target based on the proteomics result, and proved by the blocked protective effects of URA by UCHL1 inhibitor. Furthermore, hirsuteine (HTE) was identified as a potential active compound of URA binding to UCHL1 by BLI, and the binding capacity and site were verified by surface plasmon resonance (SPR) and molecular docking. CONCLUSION This work demonstrates that URA exerts effective neuroprotective activity against PD via activation of mitophagy with the involvement of UCHL1, and HTE may be a potential active compound of URA.
Collapse
Affiliation(s)
- Xingzi Hou
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Xiaomin Liang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Xin Zhao
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, PR China.
| | - Fangfang Zhuo
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Xinnuo Tong
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Xunfang Yang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Qi Zhai
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Jie Wang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Qiang Guo
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, PR China.
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| | - Qingying Zhang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, PR China.
| |
Collapse
|
24
|
Zhang Z, Wu H, Li M, Zhou F, Huang Y. From natural herbal wisdom to nano innovation: Revolutionizing tumor treatment through intervening in metabolic reprogramming. Biochim Biophys Acta Rev Cancer 2025; 1880:189263. [PMID: 39800231 DOI: 10.1016/j.bbcan.2025.189263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
In recent years, with the deepening understanding of the biological mechanisms underlying tumorigenesis, metabolic reprogramming has emerged as a pivotal process in cancer initiation, progression, and treatment resistance, gradually paving the way for new avenues in precision oncology. Natural herbal ingredients, characterized by their multi-target engagement, low toxicity, and wide-ranging biological activities, exhibit unique advantages in anti-cancer therapy. Nonetheless, the clinical application of these components has been constrained by issues such as poor solubility, low bioavailability, and inadequate stability when administered through traditional delivery methods. The advent of multifunctional nanoformulations has offered solutions to these challenges, substantially advancing the utilization of natural herbal components in precision therapy targeting tumor metabolic reprogramming. This article provides a comprehensive review of the multidimensional features of cancer metabolic reprogramming and its intricate regulatory network, highlighting the latest advancements in metabolic regulation, targeted delivery, and precision therapy achieved through natural herbs and their multifunctional nanomedicines. It also offers insights into future directions in this field. We are justified in believing that continued breakthroughs in this area will usher in safer and more effective treatment options for cancer patients, heralding a new chapter in cancer therapy.
Collapse
Affiliation(s)
- Zhengguang Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China; School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Haitao Wu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Yan Huang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| |
Collapse
|
25
|
Guo T, Hui Y, Zhu W, Ke F, Zhou T, Qiu W, Li X, Li L, Cheng H. Extended survival of a patient with gastrointestinal multiple malignancies managed with anti-PD-1 immunotherapy: a case report. Immunotherapy 2025; 17:95-101. [PMID: 39935272 PMCID: PMC11901419 DOI: 10.1080/1750743x.2025.2463309] [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: 08/28/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION The annual rise in gastrointestinal cancer cases is evident, yet the occurrence of multiple primary malignancies remains comparatively uncommon. In recent years, immunotherapy has swiftly emerged as the leading treatment for several solid tumors, including gastrointestinal cancers. Single treatments might be ineffective, necessitating the need for comprehensive integrative medicine. CASE DESCRIPTION This study reports a case of multiple cancers, including colorectal and gastric cancers. Diverse systemic treatments, like capecitabine, the combination of capecitabine and paclitaxel liposome, as well as capecitabine with toripalimab, were unsuccessful. Nevertheless, prolonged survival was attained through anti-PD-1 immunotherapy complemented by alternative medicine approaches. The patient has exceeded a 35-month survival post-initial diagnosis and 20-month survival since the subsequent diagnosis, markedly surpassing the prognosis often associated with advanced-stage multiple cancers. CONCLUSION In summary, this case underscores the potential effectiveness of a holistic, integrative medical approach in managing advanced multiple malignancies amid drug resistance and disease progression.
Collapse
Affiliation(s)
- Tianhao Guo
- Institute of Health and Regimen, Jiangsu Open University, Nanjing, Jiangsu, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yifan Hui
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wenjian Zhu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Fei Ke
- Department of Pathology, Jiangsu Provincial Hospital of Traditional Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, Jiangsu, China
| | - Tingting Zhou
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenli Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xuan Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Liu Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
26
|
Wang Y, Yuan J, Liu J, Li X, Zhou C, Qian M, Zou Z, Lu C, Huang G, Jin M. Melittin suppresses aerobic glycolysis by regulating HSF1/PDK3 to increase chemosensitivity of NSCLC. Eur J Pharmacol 2025; 986:177084. [PMID: 39547404 DOI: 10.1016/j.ejphar.2024.177084] [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: 08/12/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
Non-small cell lung cancer (NSCLC), although considered non-immunogenic, is often resistant to chemotherapy agents during the course of treatment in clinical patients. Melittin (C131H229N39O31, CAS: 20449-79-0), the major component of honey bee venom, is a promising anticancer drug. However, the mechanism employed by melittin to reverse chemotherapy resistance of NSCLC cells remains unknown. In this study, the Cell Counting Kit 8, ethynyl deoxyuridine assay, and other assays were utilized to elucidate the melittin effects upon cell proliferation. Proteomics, lung cancer (LC) tissue chip, and Western blot analysis were used to identify potential targets of melittin. A549/DDP cells were employed to investigate the melittin effects against cisplatin resistance. Also, an in vivo animal experiment was conducted to further clarify the regulatory function of melittin towards cisplatin resistance of A549/DDP cells. Results showed that melittin inhibited malignant progression of A549/DDP cells by down-regulation of pyruvate dehydrogenase kinase 3 (PDK3)-mediated aerobic glycolysis and inhibition of heat shock factor 1 (HSF1) expression. The therapeutic effect of melittin was increased by combination with KNK437 and impaired chemotherapy resistance regarding A549/DDP cells via reversing aerobic glycolysis. The in vivo experiments confirmed that melittin incremented A549/DDP cell cisplatin sensitivities. Collectively, the data suggested that melittin suppressed aerobic glycolysis by regulating HSF1/PDK3, which incremented cisplatin sensitivity of A549/DDP cells. It may provide a new treatment method for chemotherapy resistance in clinical NSCLC patients.
Collapse
Affiliation(s)
- Yuhan Wang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jiaying Yuan
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Jiao Liu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xiaodan Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Chuanqiang Zhou
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Minxuan Qian
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Zhangyan Zou
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Changlian Lu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
27
|
Wang J, He X, Jia Z, Yan A, Xiao K, Liu S, Hou M, Long Y, Ding X. Shenqi Fuzheng injection restores the sensitivity to gefitinib in non-small cell lung cancer by inhibiting the IL-22/STAT3/AKT pathway. PHARMACEUTICAL BIOLOGY 2024; 62:33-41. [PMID: 38100532 PMCID: PMC10732196 DOI: 10.1080/13880209.2023.2292266] [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: 05/27/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
CONTEXT Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. Gefitinib is a first-line treatment for NSCLC. However, its effectiveness is hindered by the development of drug resistance. At present, Shenqi Fuzheng injection (SFI) is widely accepted as an adjuvant therapy in NSCLC. OBJECTIVE This study investigates the molecular mechanism of SFI when combined with gefitinib in regulating cell progression among EGFR-TKI-resistant NSCLC. MATERIALS AND METHODS We established gefitinib-resistant PC9-GR cells by exposing gefitinib escalation from 10 nM with the indicated concentrations of SFI in PC9 cells (1, 4, and 8 mg/mL). Quantitative real-time polymerase chain reaction was performed to assess gene expression. PC9/GR and H1975 cells were treated with 50 ng/mL of interleukin (IL)-22 alone or in combination with 10 mg/mL of SFI. STAT3, p-STAT3, AKT, and p-AKT expression were evaluated using Western blot. The effects on cell proliferation, clonogenicity, and apoptosis in NSCLC cells were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation and flow cytometry assays. RESULTS SFI treatment alleviated the development of gefitinib resistance in NSCLC. PC9/GR and H1975 cells treated with SFI significantly exhibited a reduction in IL-22 protein and mRNA overexpression levels. SFI effectively counteracted the activation of the STAT3/AKT signaling pathway induced by adding exogenous IL-22 to PC9/GR and H1975 cells. Moreover, IL-22 combined with gefitinib markedly increased cell viability while reducing apoptosis. In contrast, combining SFI with gefitinib and the concurrent treatment of SFI with gefitinib and IL-22 demonstrated the opposite effect. DISCUSSION AND CONCLUSION SFI can be a valuable therapeutic option to address gefitinib resistance in NSCLC by suppressing the IL-22/STAT3/AKT pathway.
Collapse
Affiliation(s)
- Jiali Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xianhai He
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhirong Jia
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Aiwen Yan
- Jiangsu Food & Pharmaceutical Science College, Jiangsu Food Science College, Huanan, China
| | - Kang Xiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shuo Liu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengjun Hou
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yaling Long
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xuansheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Precision Medicine Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
28
|
Peng J, Li Q, Liu L, Gao P, Xing L, Chen L, Liu H, Liu Z. Exploring the material basis and molecular targets of Changma Xifeng tablet in treating Tourette syndrome: an integrative approach of network pharmacology and miRNA analysis. Metab Brain Dis 2024; 39:1573-1590. [PMID: 39436634 DOI: 10.1007/s11011-024-01408-6] [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: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 10/23/2024]
Abstract
This study was to investigate the mechanism of Changma Xifeng tablet, a traditional Chinese medicine in the treatment of Tourette syndrome. Network pharmacology was utilized to pinpoint blood-entering constituents of Changma Xifeng and explore their potential targets. Additionally, differential microRNA expression analysis was conducted to predict Tourette syndrome-associated targets, complemented by molecular docking and dynamics simulations to support the interactions of the active compounds with these targets. The study identified 98 common targets between Changma Xifeng and Tourette syndrome, which may be involved in the treatment process. A protein-protein interaction network and a drug-active ingredient-disease target network highlighted the formulation's multi-component, multi-target therapeutic approach. Eight pivotal targets-AR, GRM5, MET, RORA, HTR2A, CNR1, PDE4B, and TOP1-were identified at the intersection of microRNA and drug targets. Molecular docking revealed 12 complexes with favorable binding energies below - 7 kcal/mol, specifically: AR with Alfacalcidol, TOP1 with Albiflorin, GRM5 with Arachidic Acid, GRM5 with Palmitic Acid, AR with Arachidic Acid, AR with 2-Hydroxyoctadecanoic Acid, RORA with Pinellic Acid, RORA with Palmitic Acid, AR with Acoronene, AR with Epiacoronene, AR with 4,4'-Methylenediphenol, and HTR2A with Calycosin. Our molecular docking and molecular dynamics simulations suggest potential stable interactions between the formulation's active components and target proteins. These computational methods provide a preliminary theoretical framework that will guide our future experimental work. The study provides a scientific rationale for the use of traditional Chinese medicine in Tourette syndrome management and offers new insights for drug development.
Collapse
Affiliation(s)
- Jing Peng
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China.
| | - Qiaoling Li
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Linhui Liu
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Ping Gao
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Lipeng Xing
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Li Chen
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Hui Liu
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Zhisheng Liu
- Department of Neurology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Wan J, Xu H, Ju J, Chen Y, Zhang H, Qi L, Zhang Y, Du Z, Zhao X. Inhibition of hERG by ESEE suppresses the progression of colorectal cancer. Transl Oncol 2024; 50:102137. [PMID: 39307030 PMCID: PMC11440318 DOI: 10.1016/j.tranon.2024.102137] [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: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant cancers. Emodin is a lipophilic anthraquinone commonly found in medicinal herbs and known for its antitumor properties. However, its clinical utility has been hampered by low druggability. We designed and synthesized a new compound named Emodin succinimidyl ethyl ester (ESEE), which improves the bioavailability and preserves the original pharmacological effects of Emodin. In vitro, we have confirmed that ESEE induces apoptosis in colon cancer cells, suppresses cell proliferation, migration, and invasion, and inhibits the growth of subcutaneous transplantation tumors associated with colon cancer. And, in vivo, ESEE robustly inhibited tumor growth. Human Ether-a-go-go Related Gene (hERG) is aberrantly expressed in various cancer cells, where they play an important role in cancer progression. Focal adhesion kinase (FAK) is a tyrosine kinase overexpressed in cancer cells and plays an important role in the progression of tumors to a malignant phenotype. Mechanistically, the anti-CRC properties of ESEE are exerted through direct binding with hERG, which impedes the FAK/PI3K/AKT signaling axis-dependent apoptotic cascade.
Collapse
Affiliation(s)
- Jufeng Wan
- Department of Pharmacology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD) , (State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Haiying Xu
- Department of Pharmacology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD) , (State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Jiaming Ju
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yingjie Chen
- Department of Pharmacology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD) , (State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Hongxia Zhang
- Department of Pharmacology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD) , (State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Lingling Qi
- Department of Pharmacology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD) , (State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yan Zhang
- Department of Pharmacology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD) , (State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Institute of Clinical Pharmacy, the Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhimin Du
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China; Institute of Clinical Pharmacy, the Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Xin Zhao
- Department of Pharmacology, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD) , (State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
30
|
Jia C, Wu H, Yang A, Chen A, Wang X, Ding S, Fan B, Zhou G, Li Z, Chen J. Mechanism Research of QingReJieDu Formula for Treating Hepatitis B Virus Based on Network Pharmacology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:155915. [PMID: 39550917 DOI: 10.1016/j.phymed.2024.155915] [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: 01/29/2024] [Revised: 05/27/2024] [Accepted: 07/25/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Hepatitis B virus (HBV) is a DNA virus known to induce hepatitis and liver dysfunction, and is one of the main causes of liver cirrhosis and liver cancer. At present, there lacks a satisfactory optimal treatment plan for HBV in clinical practice, promoting the development of a novel Chinese formula, QingReJieDu Formula (QRJDF), as a potential solution. PURPOSE This study aims to explore the underlying mechanisms of QRJDF in the treatment of Hepatitis B virus (HBV) through a combination of network pharmacology and experimental validation. METHODS/STUDY DESIGN HepG2.2.15 cells were used to study the efficacy of QRJDF against HBV in vitro. Entecavir (ETV) was used as a positive control. Additionally, HBV transgenic mice served as subjects to study the in vivo efficacy of QRJDF against HBV, with serum and tissue samples analyzed post-euthanasia at 12 weeks to observe relevant indicators. UPLC-Q-TOF-MS technology was utilized to obtain the main ingredients in QRJDF. Network pharmacology was used to explore the potential ingredients and targets of QRJDF against HBV. Transcriptome sequencing was used to further explore the potential targets of QRJDF against HBV. Finally, integration of network pharmacology and transcriptomics results facilitated the screening of potential key targets and identification of potential pathways. RESULTS QRJDF demonstrated anti-HBV effects in HepG2.2.15 cells, compared to ETV control, QRJDF was more efficient in inhibiting HBV antigen levels, although it was less efficient in inhibiting HBV DNA level. In addition, the antiviral effect was verified in HBV transgenic mice. Network pharmacology results found three major active anti-HBV ingredients from QRJDF. Network pharmacology and transcriptomics revealed that QRJDF could act on the TGFβ1/Smad4 signaling pathway. CONCLUSION The study comprehensively evaluated the efficacy in vivo and in vitro, and fully confirmed that QRJDF was a potential therapeutic agent for HBV. In addition, the transcriptome technology was verified, and the key targets and approaches of QRJDF against hepatitis B were screened in combination with network pharmacology, which provided research ideas for the follow-up research of antiviral Chinese medicine.
Collapse
Affiliation(s)
- Caixia Jia
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hongxing Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Aiqing Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, P.R. 100850, China
| | - Aiping Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xueting Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, P.R. 100850, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Key Laboratory of Analytical Science and Technology of Hebei Province, College of Chemistry and Environmental Science, Hebei University, Baoding 071002, Hebei, China
| | - Shuqin Ding
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, P.R. 100850, China; College of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Baofeng Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Gangqiao Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, P.R. 100850, China.
| | - Zhihong Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing university of Chinese Medicine, No.5 Haiyuncang Hutong, Dongcheng district, Beijing 100700, China.
| | - Jianxin Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
31
|
Qiao JX, Guo DY, Tian H, Wang ZP, Fan QQ, Tian Y, Sun J, Zhang XF, Zou JB, Cheng JX, Luan F, Zhai BT. Research progress of paclitaxel nanodrug delivery system in the treatment of triple-negative breast cancer. Mater Today Bio 2024; 29:101358. [PMID: 39677523 PMCID: PMC11638641 DOI: 10.1016/j.mtbio.2024.101358] [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: 07/16/2024] [Revised: 10/27/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by the loss or low expression of estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2) and progesterone receptor (PR). Due to the lack of clear therapeutic targets, paclitaxel (PTX) is often used as a first-line standard chemotherapy drug for the treatment of high-risk and locally advanced TNBC. PTX is a diterpenoid alkaloid extracted and purified from Taxus plants, functioning as an anticancer agent by inducing and promoting tubulin polymerization, inhibiting spindle formation in cancer cells, and preventing mitosis. However, its clinical application is limited by low solubility and high toxicity. Nanodrug delivery system (NDDS) is one of the feasible methods to improve the water solubility of PTX and reduce side effects. In this review, we summarize the latest advancements in PTX-targeted NDDS, as well as its combination with other codelivery therapies for TNBC treatment. NDDS includes passive targeting, active targeting, stimuli-responsive, codelivery, and multimode strategies. These systems have good prospects in improving the bioavailability of PTX, enhancing tumor targeting, reducing toxicity, controlling drug release, and reverse tumor multidrug resistance (MDR). This review provides valuable insights into the clinical development and application of PTX-targeted NDDS in the treatment of TNBC.
Collapse
Affiliation(s)
- Jia-xin Qiao
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Department of Pharmacy, National Old Pharmacist Inheritance Studio, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Zhan-peng Wang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Qiang-qiang Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yuan Tian
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Fei Luan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Bing-tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| |
Collapse
|
32
|
Wen Y, Yi F, Zhang J, Wang Y, Zhao C, Zhao B, Wang J. Uncovering the protective mechanism of baicalin in treatment of fatty liver based on network pharmacology and cell model of NAFLD. Int Immunopharmacol 2024; 141:112954. [PMID: 39153306 DOI: 10.1016/j.intimp.2024.112954] [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: 06/12/2024] [Revised: 07/25/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Excessive nonesterified fatty acids (NEFA) impair cellular metabolism and will induce fatty liver formation in dairy cows during the periparturient. Baicalin, an active flavonoid, has great potential efficacy in alleviating lipid accumulation and ameliorating the development of fatty liver disease. Nevertheless, its mechanism remains unclear. Here, the potential mechanism of baicalin on system levels was explored using network pharmacology and in vitro experiments. Firstly, the target of baicalin and fatty liver disease was predicted, and then the protein-protein interaction (PPI) network was constructed. In addition, the Kyoto Encyclopedia of Genes and Genomes (KEGG) (q-value) pathway enrichment is performed through the Database for Annotation, Visualization, and Integrated Discovery (DAVID) server. Finally, the results of the network analysis of the in vitro treatment of bovine hepatocytes by NEFA were confirmed. The results showed that 33 relevant targets of baicalin in the treatment of liver fatty were predicted by network pharmacology, and the top 20 relevant pathways were extracted by KEGG database. Baicalin treatment can reduce triglyceride (TAG) content and lipid droplet accumulation in NEFA-treated bovine hepatocytes, and the mechanism is related to inhibiting lipid synthesis and promoting lipid oxidation. The alleviating effect of baicalin on fatty liver may be related to the up-regulation of solute vector family member 4 (SLC2A4), Down-regulated AKT serine/threonine kinase 1 (AKT1), Peroxisome proliferator-activated receptor gamma (PPARG), Epidermal growth factor receptor (EGFR), tumor necrosis factor (TNF), Interleukin 6 (IL-6) were associated. These results suggested that baicalin may modulate key inflammatory markers, and lipogenesis processes to prevent fatty liver development in dairy cows.
Collapse
Affiliation(s)
- Yongqiang Wen
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Fanxuan Yi
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Jia Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yazhou Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Chenxu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
33
|
Yang S, Lin M, Hao S, Ye H, Zhang X. Current hotspots and trends in cancer metabolic reprogramming: a scientometric analysis. Front Immunol 2024; 15:1497461. [PMID: 39588377 PMCID: PMC11586341 DOI: 10.3389/fimmu.2024.1497461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024] Open
Abstract
Background Metabolic reprogramming (MR) in cancer (CA) has been a focus of intense research in the recent two decades. This phenomenon has attracted great interest because it offers potential targets for cancer therapy. To capture the intellectual landscape of this field, we conducted a bibliometric analysis to assess the scientific output, major contributors, and trends in the MR/CA research. Methods We performed a systematic search using the Web of Science to retrieve articles published on MR of cancer from 2006 until 2023. The bibliometric tools such as Biblioshiny, VOSviewer, and Microsoft Excel were used to identify the most prolific authors, institutions, citation patterns, and keywords. We also used co-citation analysis to map the conceptual structure of the field and identify influential publications. Furthermore, we examined the literature by analyzing publication years, citations, and research impact factors. Results A total of 4,465 publications about MR/CA were retrieved. Publications on MR/CA increased rapidly from 2006 to 2023. Frontiers in Oncology published the most papers, while Cell Metabolism had the most citations. Highly cited papers were mainly published in Cancer Cell, Nature, Cell, Science and Cell Metabolism. China and the United States led the way in publications and contributed the most to MR/CA research. The University of Texas System, Chinese Academy of Sciences, and Fudan University were the most productive institutions. The profitable authors were Deberardinis Ralph J and Chiarugi Paola. The current topics included MR in tumorigenesis and progression of CA, MR of tumor cells and tumor microenvironment, the effect of MR on the CA treatment, the underlying mechanisms of MR (such as gene regulation, epigenetics, extracellular vesicles, and gut microbiota), and the modulation of MR. Some topics such as tumor microenvironment, lipid MR, circular RNA, long noncoding RNA, exosome, prognostic model, and immunotherapy may be the focus of MR/CA research in the next few years. Conclusion This study evaluated the global scientific output in the field of MR/CA research, analyzing its quantitative characteristics. It identified some significant and distinguished papers and compiled information regarding the current status and evolving trends of MR/CA research.
Collapse
Affiliation(s)
- Shanshan Yang
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Miaomiao Lin
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Shaodong Hao
- Spleen and Stomach Disease Department, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Ye
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Xuezhi Zhang
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| |
Collapse
|
34
|
Goyal H, Parwani S, Fatima K, Kaur J. Harnessing the power of Calculus bovis: Anti-cancer properties and Wnt pathway modulation in hepatocellular carcinoma. World J Gastroenterol 2024; 30:4496-4502. [DOI: 10.3748/wjg.v39.i41.4496] [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: 08/05/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024] Open
Abstract
In this manuscript, we comment on the article, which explores the anti-cancer effects of Calculus bovis (CB) in tumor biology. We highlight its potential, particularly in hepatocellular carcinoma (HCC), where it inhibits the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin pathways and induces apoptosis. CB contains compounds such as oleanolic acid and ursolic acid that target interleukin-6, mitogen-activated protein kinase 8, vascular endothelial growth factor, and caspase-3, offering anti-inflammatory and hepatoprotective benefits. The manuscript also discusses CB sativus (CBS), an artificial substitute, which has shown efficacy in reducing hepatic inflammation and oxidative stress in animal models. We emphasize the need for further research on the effects of CBS on the gut-liver axis and gut microbiota, and on targeting Wnt signaling and M2 tumor-associated macrophage as potential therapeutic strategies against HCC.
Collapse
Affiliation(s)
- Himanshi Goyal
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Sachin Parwani
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Kaneez Fatima
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
35
|
Goyal H, Parwani S, Fatima K, Kaur J. Harnessing the power of Calculus bovis: Anti-cancer properties and Wnt pathway modulation in hepatocellular carcinoma. World J Gastroenterol 2024; 30:4496-4502. [PMID: 39534422 PMCID: PMC11551676 DOI: 10.3748/wjg.v30.i41.4496] [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: 08/05/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024] Open
Abstract
In this manuscript, we comment on the article, which explores the anti-cancer effects of Calculus bovis (CB) in tumor biology. We highlight its potential, particularly in hepatocellular carcinoma (HCC), where it inhibits the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin pathways and induces apoptosis. CB contains compounds such as oleanolic acid and ursolic acid that target interleukin-6, mitogen-activated protein kinase 8, vascular endothelial growth factor, and caspase-3, offering anti-inflammatory and hepatoprotective benefits. The manuscript also discusses CB sativus (CBS), an artificial substitute, which has shown efficacy in reducing hepatic inflammation and oxidative stress in animal models. We emphasize the need for further research on the effects of CBS on the gut-liver axis and gut microbiota, and on targeting Wnt signaling and M2 tumor-associated macrophage as potential therapeutic strategies against HCC.
Collapse
Affiliation(s)
- Himanshi Goyal
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Sachin Parwani
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Kaneez Fatima
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
36
|
Wang C, Liu B, Dan W, Wei Y, Li M, Guo C, Zhang Y, Xie H. Liquiritigenin inhibits the migration, invasion, and EMT of prostate cancer through activating ER stress. Arch Biochem Biophys 2024; 761:110184. [PMID: 39447623 DOI: 10.1016/j.abb.2024.110184] [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: 07/10/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Liquiritigenin (LQ) is a monomeric compound found in licorice, a leguminous plant, and has been reported to exhibit antitumor effects in various lines of cancer cells. However, the underlying molecular mechanisms by which LQ exerts its antitumor effects remain largely unknown. In this study, the effects of LQ on the migration, invasion, and epithelial-mesenchymal transition (EMT) of prostate cancer (PCa) cells were investigated. We found that LQ effectively inhibited the migration and invasion of PCa cells in vitro, and this effect was further confirmed in xenograft lung metastasis models. In addition, LQ was found to activate endoplasmic reticulum stress (ER stress) in PCa cells. Further studies found that LQ upregulated the expression of inositol-requiring enzyme type 1α (IRE1). When IRE1 was knocked down, we observed a weakened inhibitory effect of LQ treatment on the migration and invasion of PCa cells. This observation suggests that LQ may inhibit the migration, invasion and EMT of PCa cells through activating the IRE1 branch of ER stress. In conclusion, our research may provide a novel therapeutic strategy for PCa.
Collapse
Affiliation(s)
- Chi Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Bo Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Weichao Dan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Yi Wei
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Mengxing Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Chendong Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Yishuai Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Hongjun Xie
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China.
| |
Collapse
|
37
|
Li T, Zhang Y, Li H, Zhang H, Xie J, Li Z, Zhang K, Yu Y, Mei L. Bufalin CaCO 3 Nanoparticles Triggered Pyroptosis through Calcium Overload via Na +/Ca 2+ Exchanger Reverse for Cancer Immunotherapy. NANO LETTERS 2024; 24:12691-12700. [PMID: 39347619 DOI: 10.1021/acs.nanolett.4c04061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Bufalin is a promising active ingredient in traditional Chinese medicine but has shown limited anticancer applications due to its toxicity. Here, we report BCNPs@gel, a bufalin-containing CaCO3 nanoparticle hydrogel, for enhancing cancer treatment through inducing cellular pyroptosis. Under the tumor microenvironment's low pH conditions, bufalin and Ca2+ are released from the delivery system. Bufalin serves as a direct anticancer drug and a Na+/K+-ATPase inhibitor by forcing the Na+/Ca2+ exchanger to reverse its function, which transfers Ca2+ into cytoplasm and ultimately causes Ca2+ overload-triggered pyroptosis. Meanwhile, we found that bufalin can upregulate PD-L1 in tumor cells. In combination with the PD-1 antibody, the delivery system showed a greater performance during the cancer treatment. BCNPs@gel enhances antitumor efficiency, reduces systemic side effects, extends antitumor mechanism of bufalin, and provides new strategies for inducing pyroptosis and calcium overload in cancer immunotherapy via Na+/K+-ATPase inhibitor. This work provides an application model for numerous other traditional Chinese medicine ingredients.
Collapse
Affiliation(s)
- Tingxuan Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| | - Yitong Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| | - Hanyue Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| | - Hanjie Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| | - Juntao Xie
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| | - Zimu Li
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Kai Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Yongkang Yu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, P. R. China
| |
Collapse
|
38
|
Han S, Zou J, Xiao F, Xian J, Liu Z, Li M, Luo W, Feng C, Kong N. Nanobiotechnology boosts ferroptosis: opportunities and challenges. J Nanobiotechnology 2024; 22:606. [PMID: 39379969 PMCID: PMC11460037 DOI: 10.1186/s12951-024-02842-5] [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: 06/14/2024] [Accepted: 09/07/2024] [Indexed: 10/10/2024] Open
Abstract
Ferroptosis, distinct from apoptosis, necrosis, and autophagy, is a unique type of cell death driven by iron-dependent phospholipid peroxidation. Since ferroptosis was defined in 2012, it has received widespread attention from researchers worldwide. From a biochemical perspective, the regulation of ferroptosis is strongly associated with cellular metabolism, primarily including iron metabolism, lipid metabolism, and redox metabolism. The distinctive regulatory mechanism of ferroptosis holds great potential for overcoming drug resistance-a major challenge in treating cancer. The considerable role of nanobiotechnology in disease treatment has been widely reported, but further and more systematic discussion on how nanobiotechnology enhances the therapeutic efficacy on ferroptosis-associated diseases still needs to be improved. Moreover, while the exciting therapeutic potential of ferroptosis in cancer has been relatively well summarized, its applications in other diseases, such as neurodegenerative diseases, cardiovascular and cerebrovascular diseases, and kidney disease, remain underreported. Consequently, it is necessary to fill these gaps to further complete the applications of nanobiotechnology in ferroptosis. In this review, we provide an extensive introduction to the background of ferroptosis and elaborate its regulatory network. Subsequently, we discuss the various advantages of combining nanobiotechnology with ferroptosis to enhance therapeutic efficacy and reduce the side effects of ferroptosis-associated diseases. Finally, we analyze and discuss the feasibility of nanobiotechnology and ferroptosis in improving clinical treatment outcomes based on clinical needs, as well as the current limitations and future directions of nanobiotechnology in the applications of ferroptosis, which will not only provide significant guidance for the clinical applications of ferroptosis and nanobiotechnology but also accelerate their clinical translations.
Collapse
Affiliation(s)
- Shiqi Han
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Jianhua Zou
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Jing Xian
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Ziwei Liu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Meng Li
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Wei Luo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Chan Feng
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China.
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
39
|
Xie J, Li XD, Li M, Zhu HY, Cao Y, Zhang J, Xu AJ. Advances in surface plasmon resonance for analyzing active components in traditional Chinese medicine. J Pharm Anal 2024; 14:100983. [PMID: 39411582 PMCID: PMC11474370 DOI: 10.1016/j.jpha.2024.100983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/26/2024] [Accepted: 04/23/2024] [Indexed: 10/19/2024] Open
Abstract
The surface plasmon resonance (SPR) biosensor technology is a novel optical analysis method for studying intermolecular interactions. Owing to in-depth research on traditional Chinese medicine (TCM) in recent years, comprehensive and specific identification of components and target interactions has become key yet difficult tasks. SPR has gradually been used to analyze the active components of TCM owing to its high sensitivity, strong exclusivity, large flux, and real-time monitoring capabilities. This review sought to briefly introduce the active components of TCM and the principle of SPR, and provide historical and new insights into the application of SPR in the analysis of the active components of TCM.
Collapse
Affiliation(s)
- Jing Xie
- Faculty of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xian-Deng Li
- Faculty of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Mi Li
- Faculty of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hong-Yan Zhu
- Faculty of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yan Cao
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Jian Zhang
- Faculty of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - A-Jing Xu
- Faculty of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
40
|
Wu J, Tang G, Cheng CS, Yeerken R, Chan YT, Fu Z, Zheng YC, Feng Y, Wang N. Traditional Chinese medicine for the treatment of cancers of hepatobiliary system: from clinical evidence to drug discovery. Mol Cancer 2024; 23:218. [PMID: 39354529 PMCID: PMC11443773 DOI: 10.1186/s12943-024-02136-2] [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: 08/10/2024] [Accepted: 09/20/2024] [Indexed: 10/03/2024] Open
Abstract
Hepatic, biliary, and pancreatic cancer pose significant challenges in the field of digestive system diseases due to their highly malignant nature. Traditional Chinese medicine (TCM) has gained attention as a potential therapeutic approach with long-standing use in China and well-recognized clinical benefits. In this review, we systematically summarized the clinical applications of TCM that have shown promising results in clinical trials in treating hepatic, biliary, and pancreatic cancer. We highlighted several commonly used TCM therapeutics with validated efficacy through rigorous clinical trials, including Huaier Granule, Huachansu, and Icaritin. The active compounds and their potential targets have been thoroughly elucidated to offer valuable insights into the potential of TCM for anti-cancer drug discovery. We emphasized the importance of further research to bridge the gap between TCM and modern oncology, facilitating the development of evidence-based TCM treatment for these challenging malignancies.
Collapse
Affiliation(s)
- Junyu Wu
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong
| | - Guoyi Tang
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong
| | - Chien-Shan Cheng
- Department of Digestive Endoscopy Center & Gastroenterology, Shuguang Hospital Affiliated With Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, China
| | - Ranna Yeerken
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong
| | - Yau-Tuen Chan
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong
| | - Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention &, Treatment Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yibin Feng
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong.
| | - Ning Wang
- School of Chinese Medicine, the University of Hong Kong, 3, Sasson Road, Pokfulam, Hong Kong.
| |
Collapse
|
41
|
Mo RL, Li Z, Zhang P, Sheng MH, Han GC, Sun DQ. Matrine inhibits invasion and migration of gallbladder cancer via regulating the PI3K/AKT signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8129-8143. [PMID: 38789637 DOI: 10.1007/s00210-024-03162-z] [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: 03/20/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Gallbladder cancer (GBC) is a common malignant cancer in the biliary system, which poses a serious threat to human health. It is urgent to explore ideal drugs for the treatment of GBC. Matrine is the main active ingredient of Sophora flavescentis, with a wide range of biological activities encompassing anti-inflammatory, antiviral, immunomodulatory, and anti-tumor. However, the underlying mechanism by which Matrine treats GBC is still unclear. The purpose of this study is to investigate the anti-tumor effects of Matrine on GBC in vivo and in vitro and to clarify the potential regulatory mechanisms. Here, we found that Matrine had a significant killing effect on GBC through CCK8 and flow cytometry, including arrest of cell cycle, inhibition of GBC cell, and induction of apoptosis. Further in vivo studies confirmed the inhibitory effect of Matrine on tumor growth in NOZ xenografted nude mouse. At the same time, Matrine also significantly suppressed the migration and invasion of GBC cells through scratch and Transwell experiments. In addition, by detecting the mRNA and protein levels of epithelial-mesenchymal transition (EMT) and matrix metalloproteinases, Matrine furtherly substantiated the inhibitory role on invasion and migration of GBC. From a mechanistic perspective, network pharmacology analysis suggests that the potential targets of Matrine in the treatment of GBC are enriched in the PI3K/AKT signaling pathway. Subsequently, Matrine effectively decreased the abundance of p-PI3K and p-AKT protein in vivo and in vitro. More importantly, PI3K activator (740 Y-P) antagonized the anti-tumor effect of Matrine, while PI3K inhibitor (LY294002) increased the sensitivity of Matrine for GBC. Based on the above findings, we conclude that Matrine inhibits the invasion and migration of GBC by regulating PI3K/AKT signaling pathway. Our results indicate the crucial role and regulatory mechanism of Matrine in suppressing the growth of GBC, which provides a theoretical basis for Matrine to be a candidate drug for the treatment and research of GBC.
Collapse
Affiliation(s)
- Rong-Liang Mo
- Anhui Medical University, School of Basic Medical Sciences, Hefei, 230032, China
| | - Zhuang Li
- Department of General Surgery, The Chinese People's Armed Police Forces Anhui Provincial Corps Hospital, Hefei, 230041, China
| | - Peng Zhang
- Graduate School, Anhui University of Chinese Medicine, Hefei, 230022, China
| | - Ming-Hui Sheng
- Department of General Surgery, The Chinese People's Armed Police Forces Anhui Provincial Corps Hospital, Hefei, 230041, China.
| | - Gen-Cheng Han
- Anhui Medical University, School of Basic Medical Sciences, Hefei, 230032, China.
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| | - Deng-Qun Sun
- Department of General Surgery, The Chinese People's Armed Police Forces Anhui Provincial Corps Hospital, Hefei, 230041, China.
| |
Collapse
|
42
|
Guo KC, Wang ZZ, Su XQ. Chinese Medicine in Colorectal Cancer Treatment: From Potential Targets and Mechanisms to Clinical Application. Chin J Integr Med 2024:10.1007/s11655-024-4115-8. [PMID: 39331211 DOI: 10.1007/s11655-024-4115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 09/28/2024]
Abstract
Colorectal cancer (CRC) is a global health challenge necessitating innovative therapeutic strategies. There is an increasing trend toward the clinical application of integrative Chinese medicine (CM) and Western medicine approaches. Chinese herbal monomers and formulations exert enhanced antitumor effects by modulating multiple signaling pathways in tumor cells, including inhibiting cell proliferation, inducing apoptosis, suppressing angiogenesis, reversing multidrug resistance, inhibiting metastasis, and regulating immunity. The synergistic effects of CM with chemotherapy, targeted therapy, immunotherapy, and nanovectors provide a comprehensive framework for CRC treatment. CM can mitigate drug toxicity, improve immune function, control tumor progression, alleviate clinical symptoms, and improve patients' survival and quality of life. This review summarizes the key mechanisms and therapeutic strategies of CM in CRC, highlighting its clinical significance. The potential for CM and combination with conventional treatment modalities is emphasized, providing valuable insights for future research and clinical practice.
Collapse
Affiliation(s)
- Ke-Chen Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zao-Zao Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiang-Qian Su
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
43
|
Li R, Ke H, Liu P, Yang Q, Li Y, Ke L, Wang X, Wu C, Zhang Y. Mechanisms of Yiai Fuzheng formula in the treatment of triple-negative breast cancer based on UPLC-Q-Orbitrap-HRMS, network pharmacology, and experimental validation. Heliyon 2024; 10:e36579. [PMID: 39319146 PMCID: PMC11419912 DOI: 10.1016/j.heliyon.2024.e36579] [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: 06/21/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Ethnopharmacological relevance Yiai Fuzheng formula (YAFZF), as a Traditional Chinese Medicine (TCM) prescription, has been used widely at Zhongnan Hospital of Wuhan University for its therapeutic effects and high safety on triple-negative breast cancer (TNBC). Objective In this study, we employed ultra-high-performance liquid chromatography-quadrupole/orbitrap high-resolution mass spectrometry (UPLC-Q-Orbitrap-HRMS), network pharmacology, and experimental validation to elucidate the underlying action mechanism of YAFZF in the treatment of TNBC. Methods The key active ingredients in YAFZF were analyzed using UPLC-Q-Orbitrap-HRMS, and then the potential components, target genes and signalling pathways of YAFZF were predicted using the network pharmacological method. We then used molecular docking to visualize the combination characteristics between major active components and macromolecules in the crucial pathway. In vitro experiments were conducted to investigate the inhibitory effects of YAFZF treatment on the cell viability, invasion, and migration of 4T1 and MDA-MB-231 cells. The xenograft TNBC models were constructed using female Balb/c mice, and their body weights, tumour volumes, and weights were monitored during YAFZF treatment. Quantitative real-time PCR (qRT-PCR), Hematoxylin-eosin (HE), immunohistochemistry (IHC) staining, Western blot (WB), and terminal deoxynucleotidyl transferase (TdT)-dUTP nick-end labeling (TUNEL) staining were used for further experimental validation. Results Based on UPLC-Q-Orbitrap-HRMS and network pharmacology analysis, 6 major bioactive components and 153 intersecting genes were obtained for YAFZF against TNBC. Functional enrichment analysis identified that the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) signalling pathway might be the mechanism of action of YAFZF in the treatment of TNBC. Molecular docking results suggested that the main active compounds in YAFZF had strong binding energies with the proteins in the PI3K/Akt pathway. In vitro experiments showed that YAFZF inhibited the cell viability, invasion, and migration abilities of TNBC cells. Animal experiments confirmed that YAFZF treatment suppressed tumour cell proliferation and increased apoptotic cells. PCR, HE, WB, and IHC results indicated that YAFZF could suppress xenograft tumour metastases by inhibiting the PI3K/AKT/mTOR pathway regulating the epithelial-mesenchymal transition (EMT) process. Conclusion YAFZF therapy showed its potential for reducing proliferation, invasion, and migration abilities, increasing apoptosis of TNBC cells. Furthermore, YAFZF treated TNBC by inhibiting xenograft tumour distant metastases via the regulation of EMT by the PI3K/Akt/mTOR pathway, suggesting that it may be useful as an adjuvant treatment.
Collapse
Affiliation(s)
- Ruijie Li
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Haoliang Ke
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Pan Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Qian Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yuxin Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Longzhu Ke
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiuping Wang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Chaoyan Wu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yingwen Zhang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| |
Collapse
|
44
|
Fang G, Yu W, Chen D, Ding X, Qiao L, Zhang L, Gao X, Yan Y, Huang Q, Ma J, Yin M. Development of a core outcome set of clinical research on the integration of traditional Chinese and Western medicine for spinal metastases: a study protocol. BMJ Open 2024; 14:e083315. [PMID: 39260838 PMCID: PMC11409365 DOI: 10.1136/bmjopen-2023-083315] [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: 12/17/2023] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND In recent years, the incidence of spinal metastasis (SM) has been increasing steadily. In response to this serious public health problem, researchers have made progress by using the integration of traditional Chinese and Western medicine. However, considerable heterogeneity in the definition and measurement of outcomes across clinical research studies, along with the lack of uniform measurement standards for study data, makes it difficult for researchers to compare different treatments. Therefore, it is crucial to accurately evaluate clinical research on the integration of traditional Chinese and Western medicine for SM. METHODS This study protocol outlines a comprehensive research programme based on the Core Outcome Set Standards Protocol Items. The study consists of four phases: a literature review, semistructured interviews, a two-round modified Delphi survey, a consensus meeting. Phase 1 involves a comprehensive literature review to extract outcomes used in current clinical studies of integrated traditional Chinese and Western medicine or Western medicine for the treatment of SM. A semistructured interview format will be used to survey patients and caregivers in phase 2 to collect suggestions from the patient perspective. Phase 3 involves a two-round modified Delphi survey to complete a prioritisation evaluation of outcomes to generate a candidate list for core outcome set (COS). Finally, phase 4 involves a face-to-face consensus meeting to review and establish the COS. ETHICS AND DISSEMINATION Conducted in response to the current dilemma of SM, the study was endorsed by the Spine Oncology Group of the Orthopaedic Surgeons Branch of the Chinese Physicians' Association. It will be developed and reported through a rigorous process, with the results of the study to be published in a peer-reviewed journal.Registration: COMET Registry: COMET 2938; https://www.comet-initiative.org/Studies/Details/2938.
Collapse
Affiliation(s)
| | - Wenlong Yu
- Department of Orthopedic, Longhua Hospital, Shanghai University of Traditional Chinese Medicinev, Shanghai, China
| | - Dingbang Chen
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xing Ding
- Department of Orthopedic, Longhua Hospital, Shanghai University of Traditional Chinese Medicinev, Shanghai, China
| | - Liang Qiao
- Department of Orthopedic, Longhua Hospital, Shanghai University of Traditional Chinese Medicinev, Shanghai, China
| | - Luosheng Zhang
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Gao
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yinjie Yan
- Department of Orthopedic, Longhua Hospital, Shanghai University of Traditional Chinese Medicinev, Shanghai, China
| | - Quan Huang
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Junming Ma
- Department of Orthopedic, Longhua Hospital, Shanghai University of Traditional Chinese Medicinev, Shanghai, China
| | - Mengchen Yin
- Department of Orthopedic, Longhua Hospital, Shanghai University of Traditional Chinese Medicinev, Shanghai, China
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
45
|
Zhang W, Xu Z, Du Y, Liu T, Xiong Z, Hu J, Chen L, Peng X, Zhou F. Identification of STAM-binding protein as a target for the treatment of gemcitabine resistance pancreatic cancer in a nutrient-poor microenvironment. Cell Death Dis 2024; 15:657. [PMID: 39242557 PMCID: PMC11379802 DOI: 10.1038/s41419-024-07048-z] [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: 04/25/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Pancreatic cancer (PC) is a highly malignant solid tumor whose resistance to gemcitabine (GEM) chemotherapy is a major cause of poor patient prognosis. Although PC is known to thrive on malnutrition, the mechanism underlying its chemotherapy resistance remains unclear. The current study analyzed clinical tissue sample databases using bioinformatics tools and observed significantly upregulated expression of the deubiquitinase STAMBP in PC tissues. Functional experiments revealed that STAMBP knockdown remarkably increases GEM sensitivity in PC cells. Multiple omics analyses suggested that STAMBP enhances aerobic glycolysis and suppresses mitochondrial respiration to increase GEM resistance in PC both in vitro and in vivo. STAMBP knockdown decreased PDK1 levels, an essential regulator of the aerobic glycolytic process, in several cancers. Mechanistically, STAMBP promoted the PDK1-mediated Warburg effect and chemotherapy resistance by modulating E2F1 via direct binding to E2F1 and suppressing its degradation and ubiquitination. High-throughput compound library screening using three-dimensional protein structure analysis and drug screening identified the FDA drug entrectinib as a potent GEM sensitizer and STAMBP inhibitor, augmenting the antitumor effect of GEM in a patient-derived xenograft (PDX) model. Overall, we established a novel mechanism, via the STAMBP-E2F1-PDK1 axis, by which PC cells become chemoresistant in a nutrient-poor tumor microenvironment.
Collapse
Affiliation(s)
- Wenming Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China
| | - Zheng Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China
| | - Yunyan Du
- Department of Pharmacology, Nanchang University, 461 Bayi Avenue, Nanchang, 330006, Jiangxi, PR China
| | - Tiande Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China
| | - Zhijuan Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, PR China
| | - Junwen Hu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China.
- Medical Center for Cardiovascular Diseases, Neurological Diseases and Tumors of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China.
- Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, PR China.
| | - Xiaogang Peng
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China.
| | - Fan Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China.
| |
Collapse
|
46
|
Wang D, Zhang Y, Li Q, Li Y, Li W, Zhang A, Xu J, Meng J, Tang L, Lyu S. Epigenetics: Mechanisms, potential roles, and therapeutic strategies in cancer progression. Genes Dis 2024; 11:101020. [PMID: 38988323 PMCID: PMC11233905 DOI: 10.1016/j.gendis.2023.04.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/20/2023] [Accepted: 04/14/2023] [Indexed: 07/12/2024] Open
Abstract
Mutations or abnormal expression of oncogenes and tumor suppressor genes are known to cause cancer. Recent studies have shown that epigenetic modifications are key drivers of cancer development and progression. Nevertheless, the mechanistic role of epigenetic dysregulation in the tumor microenvironment is not fully understood. Here, we reviewed the role of epigenetic modifications of cancer cells and non-cancer cells in the tumor microenvironment and recent research advances in cancer epigenetic drugs. In addition, we discussed the great potential of epigenetic combination therapies in the clinical treatment of cancer. However, there are still some challenges in the field of cancer epigenetics, such as epigenetic tumor heterogeneity, epigenetic drug heterogeneity, and crosstalk between epigenetics, proteomics, metabolomics, and other omics, which may be the focus and difficulty of cancer treatment in the future. In conclusion, epigenetic modifications in the tumor microenvironment are essential for future epigenetic drug development and the comprehensive treatment of cancer. Epigenetic combination therapy may be a novel strategy for the future clinical treatment of cancer.
Collapse
Affiliation(s)
- Dong Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yan Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qingbo Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yu Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wen Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ao Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingxuan Xu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingyan Meng
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Tang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuhua Lyu
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, China
| |
Collapse
|
47
|
Jia R, Shao S, Zhang P, Yuan Y, Rong W, An Z, Lv S, Feng Y, Liu N, Feng Q, Wang Y, Li Q. PRM1201 effectively inhibits colorectal cancer metastasis via shaping gut microbiota and short- chain fatty acids. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155795. [PMID: 38878524 DOI: 10.1016/j.phymed.2024.155795] [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: 12/03/2023] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND PRM1201 is a traditional medicine with beneficial effects against colorectal cancer (CRC) metastasis. However, the underlying mechanism of this action remains to be determined. HYPOTHESIS Remodeling microbiota and short-chain fatty acids (SCFAs) metabolism might be a potential mechanism to explain the anti-metastatic action of PRM1201, as this gut-microbiota dependent effect involves downregulation of histone deacetylation and EMT. METHODS To investigate this possibility, clinical specimens were sequenced and the correlation between the anti-metastatic efficacy of PRM1201 and the restoration of SCFA-producing bacteria was studied. To obtain solid causal evidence, a mouse metastasis model was established to detect the influence of PRM1201 on cancer metastasis. Specifically, 16S amplicon sequencing, ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) analysis, and bacterial manipulation were used to examine the gut microbiota-driven anti-metastatic action of PRM1201. RESULTS Clinical data showed that PRM1201 increased both the number of SCFA-producing bacteria and generation of SCFAs in the feces of CRC patients. A positive correlation between the anti-metastatic efficacy of PRM1201 and the restoration of SCFAs observed. The animal experiments demonstrated that PRM1201 effectively blocked CRC metastasis in a dose-dependent manner. PRM1201 treatment modulated the composition of gut microbiota, and promoted the proliferation of beneficial SCFAs producers such as Akkermansia, Lachnospiraceae_NK4A136_group and Blautia, while simultaneously reducing the abundance of pathogenic bacteria like Escherichia-Shigella. In addition, PRM1201 led to augmentation of SCFAs content. Further results indicated that the anti-cancer metastatic mechanism of PRM1201 was linked to inhibition of histone deacetylation and suppression of epithelial-to-mesenchymal transition (EMT) in metastatic lesions. Microbiota depletion treatment and fecal microbiota transplantation (FMT) underscored the microbiota-dependent nature of this phenomenon. Moreover, this anti-colorectal cancer metastatic effect and mechanism of total SCFAs and single SCFA were also confirmed. CONCLUSION In summary, PRM1201 exerts its anti-metastatic effects by modulating SCFA-producing bacteria and enhancing the production of SCFAs. Furthermore, the prebiotic-like actions of PRM1201, along with the PRM1201-treated bacteria, function as inhibitors of histone deacetylases (DHACs) thereby effectively suppressing EMT events.
Collapse
Affiliation(s)
- Ru Jia
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shiyun Shao
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pingping Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuan Yuan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenqing Rong
- Department of Medical Oncology, Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Ziming An
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai 201203, China
| | - Sheng Lv
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai 201203, China
| | - Yuanyuan Feng
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ningning Liu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qin Feng
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Institute of Liver Diseases, Shanghai 201203, China; Central Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai 201203, China.
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qi Li
- Department of Chinese Medicine & Integrative Medicine, Shanghai Geriatric Medical Center, Zhongshan Hospital, Fudan University, 2560 Chunshen Road, Shanghai 201104, China.
| |
Collapse
|
48
|
Yan Z, Li Y, Xia T, Wang K, Liao Z, Zhang L, Wang Y, Shen P, Bai Z, Wang N, Zhou W, Ni Z, Dou Y, Gao Y. Revitalizing gut health: Liangxue guyuan yishen decoction promotes akkermansia muciniphila -induced intestinal stem cell recovery post-radiation in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155888. [PMID: 39084128 DOI: 10.1016/j.phymed.2024.155888] [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: 04/27/2024] [Revised: 06/28/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND The efficacy of Liangxue Guyuan Yishen Decoction (LGYD), a traditional Chinese medicine, has been scientifically proven in the treatment of radiation-induced intestinal injury (RIII) and preservation of intestinal integrity and function following high-dose radiation exposure. However, further investigation is required to comprehensively elucidate the precise mechanisms underlying the therapeutic effects of LGYD in order to provide potential pharmaceutical options for radiation protection. PURPOSE This study aims to elucidate the potential mechanism through which LGYD exerts its therapeutic effects on RIII by modulating the gut microbiota (GM). METHODS 16 s rRNA analysis was employed to assess the impact of varying doses of whole body irradiation (WBI) on GM in order to establish an appropriate model for this study. The effects of LGYD on GM and SCFA were evaluated using 16 s rRNA and Quantification of SCFA. UHPLC-QE-MS was utilized to identify the active components in LGYD as well as LGYD drug containing serum (LGYD-DS). Subsequently, immunofluorescence and immunohistochemical staining were conducted to validate the influence of LGYD and/or characteristic microbiota on RIII recovery in vivo. The effects of LGYD-DS, characteristic flora, and SCFA on intestinal stem cell (ISC) were assessed by measuring organoid surface area in intestinal organoid model. RESULTS The species composition and abundance of GM were significantly influenced by whole-body irradiation with a dose of 8.5 Gy, which was used as in vivo model. LGYD significantly improves the survival rate and promotes recovery from RIII. Additionally, LGYD exhibited a notable increase in the abundance of Akkermansia muciniphila (AKK) and levels of SCFA, particularly isobutyric acid. LGYD-DS consisted of seven main components derived from herbs of LGYD. In vivo experiments indicated that both LGYD and AKK substantially enhanced the survival rate after radiation and facilitated the recovery process for intestinal structure and function. In the organoid model, treatment with LGYD-DS, AKK supernatant or isobutyric acid significantly increased organoid surface area. CONCLUSIONS LGYD has the potential to enhance RIII by promoting the restoration of intestinal stem cell, which is closely associated with the upregulation of AKK abundance and production of SCFA, particularly isobutyric acid.
Collapse
Affiliation(s)
- Ziqiao Yan
- Department of Traditional Chinese Medicine, the Sixth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China; Chinese PLA Medical School, Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Yangshuo Li
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, PR China
| | - Tiantian Xia
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China; Medical College of Qinghai University, Xining, PR China
| | - Kaili Wang
- Department of Traditional Chinese Medicine, the Sixth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China
| | - Zebin Liao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Liangliang Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Yuguo Wang
- Department of Traditional Chinese Medicine, the Sixth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China
| | - Pan Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Zhijie Bai
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Ningning Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Zhexin Ni
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China.
| | - Yongqi Dou
- Department of Traditional Chinese Medicine, the Sixth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China; Chinese PLA Medical School, Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China.
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China; Medical College of Qinghai University, Xining, PR China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, PR China; State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, PR China.
| |
Collapse
|
49
|
Xiao X, Li Z, Li Q, Qing L, Wang Y, Ye F, Dong Y, Di X, Mi J. Exploring the clinical and biological significance of the cell cycle-related gene CHMP4C in prostate cancer. BMC Med Genomics 2024; 17:210. [PMID: 39138470 PMCID: PMC11323463 DOI: 10.1186/s12920-024-01970-z] [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: 09/10/2023] [Accepted: 07/25/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) stands as the second most prevalent malignancy impacting male health, and the disease's evolutionary course presents formidable challenges in the context of patient treatment and prognostic management. Charged multivesicular body protein 4 C (CHMP4C) participates in the development of several cancers by regulating cell cycle functions. However, the role of CHMP4C in prostate cancer remains unclear. METHODS In terms of bioinformatics, multiple PCa datasets were employed to scrutinize the expression of CHMP4C. Survival analysis coupled with a nomogram approach was employed to probe into the prognostic significance of CHMP4C. Gene set enrichment analysis (GSEA) was conducted to interrogate the functional implications of CHMP4C. In terms of cellular experimentation, the verification of RNA and protein expression levels was executed through the utilization of qRT-PCR and Western blotting. Upon the establishment of a cell line featuring stable CHMP4C knockdown, a battery of assays, including Cell Counting Kit-8 (CCK-8), wound healing, Transwell, and flow cytometry, were employed to discern the impact of CHMP4C on the proliferation, migration, invasion, and cell cycle function of PCa cells. RESULTS The expression of CHMP4C exhibited upregulation in both PCa cells and tissues, and patients demonstrating elevated CHMP4C expression levels experienced a notably inferior prognosis. The nomogram, constructed using CHMP4C along with clinicopathological features, demonstrated a commendable capacity for prognostic prediction. CHMP4C knockdown significantly inhibited the proliferation, migration, and invasion of PCa cells (LNcaP and PC3). CHMP4C could impact the advancement of the PCa cell cycle, and its expression might be regulated by berberine. Divergent CHMP4C expression among PCa patients could induce alterations in immune cell infiltration and gene mutation frequency. CONCLUSIONS Our findings suggest that CHMP4C might be a prognostic biomarker in PCa, potentially offering novel perspectives for the advancement of precision therapy for PCa.
Collapse
Affiliation(s)
- Xi Xiao
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zonglin Li
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Qingchao Li
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Liangliang Qing
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Yanan Wang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Fuxiang Ye
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Yajia Dong
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xiaoyu Di
- Department of plastic surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Jun Mi
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
50
|
Pan YF, Zhong L, Wang M, Jiang TY, Lin YK, Chen YB, Li X, Hu HP, Zhou HB, Yan HZ, Dong LW. PTEN status determines therapeutic vulnerability to celastrol in cholangiocarcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155790. [PMID: 38851099 DOI: 10.1016/j.phymed.2024.155790] [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: 03/20/2024] [Revised: 05/15/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND A balanced protein homeostasis network helps cholangiocarcinoma (CCA) maintain their oncogenic growth, and disrupting proteostasis therapeutically will induce proteotoxic stress. Phosphatase and tensin homolog (PTEN) have been reported to be involved in proteostasis, and PTEN-associated pathways are commonly altered in CCA. Celastrol, a triterpene from plants, exhibits cytotoxic effects in various types of cancer. However, the underlying mechanisms remain unclear. PURPOSE We investigated the therapeutic effect of celastrol in CCA and identified the molecular characteristics of tumors that were sensitive to celastrol. The target of celastrol was explored. We then evaluated the candidate combination therapeutic strategy to increase the effectiveness of celastrol in celastrol-insensitive CCA tumors. METHODS Various CCA cells were categorized as either celastrol-sensitive or celastrol-insensitive based on their response to celastrol. The molecular characteristics of cells from different groups were determined by RNA-seq. PTEN status and its role in proteasome activity in CCA cells were investigated. The CMAP analysis, molecular docking, and functional assay were performed to explore the effect of celastrol on proteasome activities. The correlation between PTEN status and clinical outcomes, as well as proteasomal activity, were measured in CCA patients. The synergistic therapeutic effect of autophagy inhibitors on celastrol-insensitive CCA cells were measured. RESULTS Diverse responses to celastrol were observed in CCA cells. PTEN expression varied among different CCA cells, and its status could impact cell sensitivity to celastrol: PTENhigh tumor cells were resistant to celastrol, while PTENlow cells were more sensitive. Celastrol induced proteasomal dysregulation in CCA cells by directly targeting PSMB5. Cells with low PTEN status transcriptionally promoted proteasome subunit expression in an AKT-dependent manner, making these cells more reliant on proteasomal activities to maintain proteostasis. This caused the PTENlow CCA cells sensitive to celastrol. A negative correlation was found between PTEN levels and the proteasome signature in CCA patients. Moreover, celastrol treatment could induce autophagy in PTENhigh CCA cells. Disrupting the autophagic pathway in PTENhigh CCA cells enhanced the cytotoxic effect of celastrol. CONCLUSION PTEN status in CCA cells determines their sensitivity to celastrol, and autophagy inhibitors could enhance the anti-tumor effect in PTENhigh CCA.
Collapse
Affiliation(s)
- Yu-Fei Pan
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education. 225 Changhai Road, Shanghai 200438, China
| | - Lin Zhong
- Department of Pathology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine (TCM), 358 Datong Road, Shanghai, 200137, China
| | - Min Wang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 700 Moyu Road, Shanghai, 201805, China
| | - Tian-Yi Jiang
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education. 225 Changhai Road, Shanghai 200438, China
| | - Yun-Kai Lin
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
| | - Yi-Bin Chen
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China
| | - Xin Li
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education. 225 Changhai Road, Shanghai 200438, China
| | - He-Ping Hu
- Department of Hepatobiliary Medicine, Eastern Hepatobiliary Surgery Hospital, 700 Moyu Road, Shanghai, 201805, China
| | - Hua-Bang Zhou
- Department of Hepatobiliary Medicine, Eastern Hepatobiliary Surgery Hospital, 700 Moyu Road, Shanghai, 201805, China.
| | - Hong-Zhu Yan
- Department of Pathology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine (TCM), 358 Datong Road, Shanghai, 200137, China.
| | - Li-Wei Dong
- National Center for Liver Cancer, Naval Medical University, 366 Qianju Road, Shanghai, 201805, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer (SMMU), Ministry of Education. 225 Changhai Road, Shanghai 200438, China.
| |
Collapse
|