1
|
Shen P, Wu S, Chen Y, Feng G, Guo X, Chen Y, Wang Z, Shen Y, Wang H, Li K. Yi Gong San inhibits tumor immune escape by sensitizing colorectal cancer stem cells via the NF-κB pathway. Hereditas 2025; 162:64. [PMID: 40247422 PMCID: PMC12004741 DOI: 10.1186/s41065-025-00412-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE Colorectal cancer (CRC), as a highly prevalent malignant tumor globally, faces the dual challenges of drug resistance of cancer stem cells and immune escape in its treatment. Although the traditional Chinese medicine Yigong San (YGS) shows potential in improving the clinical adverse reactions of CRC, its core active components and mechanism of action remain unclear. Based on network pharmacology screening, this study for the first time discovered that Gomisin B might regulate the progression of CRC through the Toll-like receptor 4/Nuclear Factor-kappa B (TLR4/NF-κB) signaling pathway, and aimed to systematically reveal the molecular mechanisms by which YGS and Gomisin B inhibited the malignant phenotypes and immune escape of CRC cells. METHODS The The Cancer Genome Atlas (TCGA) database was integrated with network pharmacology analysis to screen for the key target of CRC, Gomisin B, and its associated TLR4/NF-κB pathway. Through in vitro CRC stem cell models and mouse xenograft tumor models, techniques such as CCK-8, Transwell, flow cytometry, qPCR/WB were used to evaluate the effects of YGS and Gomisin B on proliferation, migration, invasion, apoptosis, and epithelial-mesenchymal transition (EMT), and to detect the expression of TLR4 and downstream inflammatory factors. RESULTS Both YGS and Gomisin B inhibited the proliferation, migration and invasion of CRC stem cells and tumor tissues. Meanwhile, they promoted apoptosis but reduced the expression of the inflammatory factor TLR4 and proteins associated with the NF-κB pathway, thereby exerting suppressive effects on tumorigenesis and disease progression. YGS might also impede EMT progression through modulation of the NF-κB pathway. CONCLUSION This study for the first time elucidated the dual anti-tumor mechanisms of YGS, which sensitized CRC stem cells and inhibited immune escape by targeting the TLR4/NF-κB pathway through Gomisin B. It provides a pharmacological basis for the modern research of traditional Chinese medicine compound against CRC. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Peng Shen
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Shunli Wu
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Yi Chen
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Guangjing Feng
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Xue Guo
- Infection Control Department, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Yingguo Chen
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Zhigang Wang
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China
| | - Youfeng Shen
- Department of Laboratory Medicine, Chongqing Precision Medical Industry Technology Research Institute, Chongqing, China
| | - Hongbo Wang
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China.
| | - Ke Li
- Department of General Surgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400000, China.
| |
Collapse
|
2
|
Feng F, Hu P, Chen J, Peng L, Wang L, Tao X, Lian C. Mechanism research on inhibition of gastric cancer in vitro by the extract of Pinellia ternata based on network pharmacology and cellular metabolomics. Open Med (Wars) 2025; 20:20241131. [PMID: 39989612 PMCID: PMC11843166 DOI: 10.1515/med-2024-1131] [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: 05/25/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 02/25/2025] Open
Abstract
Background and purpose Gastric cancer is a kind of malignant tumor with high incidence and high mortality, which has strong tumor heterogeneity. A classic Chinese medicine, Pinellia ternata (PT), was shown to exert therapeutic effects on gastric cancer cells. However, its chemical and pharmacological profiles remain to be elucidated. In the current study, we aimed to reveal the mechanism of PT in treating gastric cancer cells through metabolomic analysis and network pharmacology. Methods Metabolomic analysis of two strains of gastric cancer cells treated with the Pinellia ternata extract (PTE) was used to identify differential metabolites, and the metabolic pathways were enriched by MetaboAnalyst. Then, network pharmacology was applied to dig out the potential targets against gastric cancer cells induced by PTE. The integrated network of metabolomics and network pharmacology was constructed based on Cytoscape. Results The PTE was confirmed to significantly inhibit cell proliferation, migration, and invasion of HGC-27 and BGC-823 cells. The results of cellular metabolomics showed that 61 metabolites were differently expressed in gastric cancer cells of the experimental and control groups. Through pathway enrichment analysis, 16 metabolites were found to be involved in the glycerophospholipid metabolism, purine metabolism, sphingolipid metabolism, and tryptophan metabolism. Combined with network pharmacology, seven bioactive compounds were found in PT, and the networks of bioactive compound-target gene-metabolic enzyme-metabolite interactions were constructed. Conclusions In conclusion, this study revealed the complicated mechanisms of PT against gastric cancer. Our work provides a novel paradigm to identify the potential mechanisms of pharmacological effects derived from a natural compound.
Collapse
Affiliation(s)
- Fan Feng
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Ping Hu
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Jun Chen
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Lei Peng
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Luyao Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China
| | - Xingkui Tao
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China
| |
Collapse
|
3
|
Wen S, Chang S, Zhang H, Zhang W, Guo Y, Zhang N, Yang A, Sun Y, Liu Z. Shikonin modulates activated fibroblast apoptosis in silicosis fibrosis via the PI3K/Akt signaling pathway: A network pharmacology approach. Toxicol Appl Pharmacol 2025; 495:117236. [PMID: 39855310 DOI: 10.1016/j.taap.2025.117236] [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: 11/18/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Silicosis is a lung disease caused by the inhalation of free crystalline silica and is characterized mainly by lung inflammation and progressive pulmonary fibrosis. Shikonin, a biologically active compound isolated from the traditional Chinese medicine Comfrey, has been shown to have significant antifibrotic effects. However, the molecular mechanisms underlying the antifibrotic effects of SHK in silicosis remain unclear. METHODS This study used a combination of network pharmacology, molecular docking, molecular dynamics simulation, and in vitro experimental validation to investigate the potential targets of SHK in silicosis. RESULTS Network pharmacology analysis identified 208 cross genes associated with disease drugs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis show that these intersecting genes are significantly associated with the PI3K/Akt signaling pathway. Protein protein interaction (PPI) network analysis further revealed 10 core crossover genes, namely ALB, Akt1, STAT3, CASP3, EGFR, MMP9, Bcl-2, ESR1, HSP90AA1, and NF-κB1. Among them, Akt1 and Bcl-2 have the strongest binding ability to SHK. The in vitro experimental results showed that SHK can significantly inhibit the activation of fibroblasts and promote apoptosis of activated fibroblasts through the PI3K/Akt signaling pathway. CONCLUSION SHK alleviates silica induced silicosis fibrosis by inhibiting the transformation of fibroblasts into myofibroblasts through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Shengpeng Wen
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Sirong Chang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Huning Zhang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Wenyue Zhang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Yi Guo
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Na Zhang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Anning Yang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China.
| | - Yue Sun
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China.
| | - Zhihong Liu
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China.
| |
Collapse
|
4
|
Pan Y, Ma T, Chen D, Wang Y, Peng Y, Lu T, Yin X, Li H, Zhang G, Wang X. Scutellaria barbata D.Don and Scleromitrion diffusum (Willd.) R.J.Wang inhibits the progression of triple negative breast cancer though the activation inhibition of NF-κB triggered by CAFs-derived IL6. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118656. [PMID: 39121924 DOI: 10.1016/j.jep.2024.118656] [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: 05/27/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The treatment options for triple-negative breast cancer (TNBC) are limited. Traditional Chinese Medicine (TCM) plays an important role in the treatment of TNBC. The herb pair Scutellaria barbata D.Don and Scleromitrion diffusum (Willd.) R.J.Wang (SH) is commonly used in clinical practice for its anti-tumor properties. It has been proven to have good therapeutic effects on tumor-related diseases, but the underlying molecular mechanisms are not yet fully explained. AIM OF STUDY Through bioinformatics, it was validated that IL6, primarily derived from cancer-associated fibroblasts (CAFs), is associated with poor prognosis. Additionally, cell and animal experiments confirmed that SH inhibits tumor proliferation, migration, and growth in an orthotopic tumor model by suppressing the IL6/NF-κB pathway. MATERIALS AND METHODS GEO, TCGA and HPA databases were used to analyze the prognostic value of CAFs and IL6, then IL6 resource was detected. After the bioinformatics, the influence of CAFs and CAFs-derived IL6 on TNBC was verified by experiments both in vitro and in vivo. Cell clone formation assay, wound-Healing assay, and Transwell assay were used to detect the promotion of CAFs and CAFs-derived IL6 and the inhibition of SH in vitro. TNBC model in mice was used to prove the promotion of CAFs and CAFs-derived IL6 and the inhibition of SH in vivo. The biological pathway of NF-κB was explored by western blotting through detecting unique molecules. RESULTS Bioinformatics analysis revealed that higher proportion of CAFs and elevated level of IL6 were significantly associated with poor prognosis in TNBC. At the same time, IL6 was proved predominantly derived from CAFs. After the indication of bioinformatics, experiments in vitro demonstrated that both CAFs and IL6 could enhance the clone formation and migration ability of MDA-MD-231 cells (231), furthermore, the promotion of CAFs was related with the level of IL6. Based on these data, mechanism was detected that CAFs-derived IL6 enhancement was closely related to the activation of NF-κB signaling pathway, while the activation can be reduced by SH. In the end, the promotion of CAFs/CAFs-derived IL6/NF-κB and the efficacy of SH inhibition were both confirmed by experiments in vivo. CONCLUSIONS Bioinformatics data indicates that higher proportion of CAFs and higher level of CAFs-derived IL6 are significantly related to poorer survival of TNBC. CAFs and CAFs-derived IL6 were proved to promote the progression of TNBC both in vitro and in vivo, and the process of which was significantly related to the activation of NF-κB. SH inhibited the progress of TNBC, which was proved to be closely related to CAFs/CAFs-derived IL6/NF-κB.
Collapse
Affiliation(s)
- Yuancan Pan
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Tingting Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Dong Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Yue Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu Peng
- Shandong University of Traditional Chinese Medicine, Shandong, 250355, China
| | - Taicheng Lu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Xiaohui Yin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Haiming Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Ganlin Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Xiaomin Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| |
Collapse
|
5
|
Wang Z, Zhang Z, Azami NLB, Hui D, Wang Z, Xie D, Ye G, Liu N, Sun M. An Integrated Approach Using Network Pharmacology and Experimental Validation to Reveal the Therapeutic Mechanism of Weifuchun in Treating Gastric Cancer. J Med Food 2024; 27:1168-1182. [PMID: 39142714 DOI: 10.1089/jmf.2024.k.0126] [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] [Indexed: 08/16/2024] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy affecting the gastrointestinal tract. Weifuchun (WFC), a Chinese herbal prescription comprising red ginseng, Isodon amethystoides, and Fructus aurantii, is widely used in China for various chronic stomach disorders. However, its therapeutic role and mechanisms in treating GC remain unexplored. In a randomized, controlled, single-blind trial involving postoperative stages II and III GC patients, we compared adjuvant chemotherapy plus WFC (chemo plus WFC group) to adjuvant chemotherapy alone (chemo group) over 6 months. We assessed recurrence and metastasis rates and used systematic pharmacology to predict WFC's active components, screen target genes, and construct network interaction maps, were validated through in vitro experiments. The combined therapy significantly reduced 2-year recurrence and metastasis rates. We identified 67 active ingredients, 211 drug target proteins, 1539 disease targets, 105 shared targets, and 188 signaling pathways associated with WFC. WFC impacted cell apoptosis, proliferation, and the inflammatory response, with top tumor-related signaling pathways involving 5'-adenosine monophosphate-activated protein kinase (AMPK), mitogen-activated protein kinase, nuclear factor kappa-B (NFKB), and apoptosis. In vitro, WFC inhibited proliferation and migration while inducing apoptosis in GC cells, reduced VEGFA, TNFa, and IL6 expressions. Immunocytochemistry showed increased p-AMPK staining, and molecular analysis revealed decreased NFKB and phosphorylation of extracellular-regulated protein kinase 1/2 (ERK1/2) levels, increased p-AMPK and BAX protein levels in WFC-treated cells, effects reversed by Compound C. WFC's antitumor effects involve AMPK-dependent ERK1/2 and NFKB pathways, regulating proliferation, migration, and apoptosis in GC cells.
Collapse
Affiliation(s)
- Ziyuan Wang
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pathology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhipeng Zhang
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Institute of Oncology, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Nisma Lena Bahaji Azami
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dengcheng Hui
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheng Wang
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong Xie
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guan Ye
- Central Research Institute of Shanghai Pharmaceutical Group Co, Ltd, Shanghai, China
| | - Ningning Liu
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingyu Sun
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Feng F, Hu P, Peng L, Xu L, Chen J, Chen Q, Zhang X, Tao X. Integrated network pharmacology and metabolomics to reveal the mechanism of Pinellia ternata inhibiting non-small cell lung cancer cells. BMC Complement Med Ther 2024; 24:263. [PMID: 38992647 PMCID: PMC11238457 DOI: 10.1186/s12906-024-04574-3] [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: 09/13/2023] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
Lung cancer is a malignant tumor with highly heterogeneous characteristics. A classic Chinese medicine, Pinellia ternata (PT), was shown to exert therapeutic effects on lung cancer cells. However, its chemical and pharmacological profiles are not yet understood. In the present study, we aimed to reveal the mechanism of PT in treating lung cancer cells through metabolomics and network pharmacology. Metabolomic analysis of two strains of lung cancer cells treated with Pinellia ternata extracts (PTE) was used to identify differentially abundant metabolites, and the metabolic pathways associated with the DEGs were identified by MetaboAnalyst. Then, network pharmacology was applied to identify potential targets against PTE-induced lung cancer cells. The integrated network of metabolomics and network pharmacology was constructed based on Cytoscape. PTE obviously inhibited the proliferation, migration and invasion of A549 and NCI-H460 cells. The results of the cellular metabolomics analysis showed that 30 metabolites were differentially expressed in the lung cancer cells of the experimental and control groups. Through pathway enrichment analysis, 5 metabolites were found to be involved in purine metabolism, riboflavin metabolism and the pentose phosphate pathway, including D-ribose 5-phosphate, xanthosine, 5-amino-4-imidazolecarboxyamide, flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). Combined with network pharmacology, 11 bioactive compounds were found in PT, and networks of bioactive compound-target gene-metabolic enzyme-metabolite interactions were constructed. In conclusion, this study revealed the complicated mechanisms of PT against lung cancer. Our work provides a novel paradigm for identifying the potential mechanisms underlying the pharmacological effects of natural compounds.
Collapse
Affiliation(s)
- Fan Feng
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Ping Hu
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Lei Peng
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Lisheng Xu
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Jun Chen
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Qiong Chen
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Xingtao Zhang
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Xingkui Tao
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China.
| |
Collapse
|
7
|
Luo Y, Zhang G, Hu C, Huang L, Wang D, Chen Z, Wang Y. The Role of Natural Products from Herbal Medicine in TLR4 Signaling for Colorectal Cancer Treatment. Molecules 2024; 29:2727. [PMID: 38930793 PMCID: PMC11206024 DOI: 10.3390/molecules29122727] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The toll-like receptor 4 (TLR4) signaling pathway constitutes an intricate network of protein interactions primarily involved in inflammation and cancer. This pathway triggers intracellular signaling cascades, modulating transcription factors that regulate gene expression related to immunity and malignancy. Previous studies showed that colon cancer patients with low TLR4 expression exhibit extended survival times and the TLR4 signaling pathway holds a significant role in CRC pathogenesis. In recent years, traditional Chinese medicines (TCMs) have garnered substantial attention as an alternative therapeutic modality for CRC, primarily due to their multifaceted composition and ability to target multiple pathways. Emerging evidence indicates that specific TCM products, such as andrographolide, rosmarinic acid, baicalin, etc., have the potential to impede CRC development through the TLR4 signaling pathway. Here, we review the role and biochemical processes of the TLR4 signaling pathway in CRC, and natural products from TCMs affecting the TLR4 pathway. This review sheds light on potential treatment strategies utilizing natural TLR4 inhibitors for CRC, which contributes to the advancement of research and accelerates their clinical integration into CRC treatment.
Collapse
Affiliation(s)
- Yan Luo
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| | - Guochen Zhang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| | - Chao Hu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Lijun Huang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| | - Dong Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| | - Zhejie Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yumei Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| |
Collapse
|
8
|
Sun J, Wei Y, Wang J, Hou M, Su L. Treatment of colorectal cancer by traditional Chinese medicine: prevention and treatment mechanisms. Front Pharmacol 2024; 15:1377592. [PMID: 38783955 PMCID: PMC11112518 DOI: 10.3389/fphar.2024.1377592] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Colorectal cancer (CRC) is a significant global health burden, with high morbidity and mortality rates. It is often diagnosed at middle to advanced stage, affecting approximately 35% of patients at the time of diagnosis. Currently, chemotherapy has been used to improve patient prognosis and increase overall survival. However, chemotherapy can also have cytotoxic effects and lead to adverse reactions, such as inhibiting bone marrow hematopoiesis, causing digestive dysfunction, hand-foot syndrome, and even life-threatening conditions. In response to these adverse effects, researchers have proposed using Traditional Chinese Medicine (TCM) as an option to treat cancer. TCM research focuses on prescriptions, herbs, and components, which form essential components of the current research in Chinese medicine. The study and implementation of TCM prescriptions and herbs demonstrate its distinctive holistic approach to therapy, characterized by applying multi-component and multi-target treatment. TMC components have advantages in developing new drugs as they consist of single ingredients, require smaller medication dosages, have a precise measure of pharmacodynamic effects, and have a clear mechanism of action compared to TCM prescriptions and herbs. However, further research is still needed to determine whether TMC components can fully substitute the therapeutic efficacy of TCM prescriptions. This paper presents a comprehensive analysis of the research advancements made in TCM prescriptions, herbs, and components. The findings of this study can serve as a theoretical basis for researchers who are interested in exploring the potential of TCM for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Jiaxin Sun
- Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, Inner Mongolia, China
| | - Ying Wei
- Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, Inner Mongolia, China
| | - Jia Wang
- Department of Gynaecology, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
| | - Mingxing Hou
- Department of Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Liya Su
- Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, Inner Mongolia, China
| |
Collapse
|
9
|
Meng FD, Yuan L, Lu DD, Yang YT, Xu DJ, Che MY, Nan Y. Anti-tumor effect of coix seed based on the theory of medicinal and food homology. World J Clin Oncol 2023; 14:593-605. [PMID: 38179404 PMCID: PMC10762529 DOI: 10.5306/wjco.v14.i12.593] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 10/26/2023] [Accepted: 11/24/2023] [Indexed: 12/22/2023] Open
Abstract
Coix seed is a dry and mature seed of Coix lacryma-jobi L.var.ma-yuen (Roman.) Stapf in the Gramineae family. Coix seed has a sweet, light taste, and a cool nature. Coix seed enters the spleen, stomach, and lung meridians. It has the effects of promoting diuresis and dampness, strengthening the spleen to prevent diarrhea, removing arthralgia, expelling pus, and detoxifying and dispersing nodules. It is used for the treatment of edema, athlete's foot, poor urination, spleen deficiency and diarrhea, dampness and obstruction, lung carbuncle, intestinal carbuncle, verruca, and cancer. The medicinal and health value is high, and it has been included in the list of medicinal and food sources in China, which has a large development and application space. This article reviews the current research achievements in the processing methods and anti-tumor activities of Coix seed and provides examples of its clinical application in ancient and modern times, aiming to provide reference for further research on Coix seed and contribute to its clinical application and development. Through the analysis of the traditional Chinese patent medicines, and simple preparations and related health food of Coix seed queried by Yaozhi.com, the source, function, and dosage form of Coix seed were comprehensively analyzed, with a view of providing a reference for the development of Coix seed medicine and food.
Collapse
Affiliation(s)
- Fan-Di Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Dou-Dou Lu
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ya-Ting Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Duo-Jie Xu
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Meng-Ying Che
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
10
|
Zhu X, Jiang C, Wang Z, Zhu X, Yuan F, Yang Y. PSKH1 affects proliferation and invasion of osteosarcoma cells via the p38/MAPK signaling pathway. Oncol Lett 2023; 25:144. [PMID: 36936027 PMCID: PMC10018237 DOI: 10.3892/ol.2023.13730] [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: 09/15/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Malignant osteosarcoma (OS) is a tumor of bone and soft tissue that metastasizes early and has a high mortality rate. Protein serine kinase H1 (PSKH1), an autophosphorylating human protein serine kinase, controls the trafficking of serine/arginine-rich domain, with downstream effects on mRNA processing. It is also associated with tumor progression. However, how this protein contributes to OS progression and metastasis is unknown. The present study evaluated the potential effect of PSKH1 on proliferation of human OS cells. OS cell lines were used in Cell Counting Kit-8, colony formation, wound-healing and Transwell assays, to investigate cellular processes such as proliferation, migration and invasion and underlying molecular mechanisms. Expression of PSKH1 in OS tissue was significantly greater than in adjacent non-malignant tissue. PSKH1 knockdown inhibited the proliferation, migration and invasion of OS cells. Conversely, PSKH1 overexpression promoted proliferation of OS cells. PSKH1 upregulated phosphorylated-p38 in OS cells. Moreover, the p38 MAPK inhibitor SB203580 effectively blocked the tumor-promoting action of PSKH1. Furthermore, PSKH1 knockdown inhibited tumor growth and metastasis in vivo. In conclusion, these findings suggested that PSKH1 promoted OS proliferation, migration and invasion. Thus, PSKH1 may serve an oncogenic role in the development of human OS.
Collapse
Affiliation(s)
- Xingfei Zhu
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Chao Jiang
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Zhiyuang Wang
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Xiaozhong Zhu
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Feng Yuan
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Yi Yang
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
- Correspondence to: Dr Yi Yang, Department of Orthopedics, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, P.R. China, E-mail:
| |
Collapse
|