101
|
Pawar K, Gupta PP, Solanki PS, Niraj RRK, Kothari SL. Targeting SLC4A4: A Novel Approach in Colorectal Cancer Drug Repurposing. Curr Issues Mol Biol 2025; 47:67. [PMID: 39852182 PMCID: PMC11764095 DOI: 10.3390/cimb47010067] [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: 12/23/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a complex and increasingly prevalent malignancy with significant challenges in its treatment and prognosis. This study aims to explore the role of the SLC4A4 transporter as a biomarker in CRC progression and its potential as a therapeutic target, particularly in relation to tumor acidity and immune response. METHODS The study utilized computational approaches, including receptor-based virtual screening and high-throughput docking, to identify potential SLC4A4 inhibitors. A model of the human SLC4A4 structure was generated based on CryoEM data (PDB ID 6CAA), and drug candidates from the DrugBank database were evaluated using two computational tools (DrugRep and CB-DOCK2). RESULTS The study identified the compound (5R)-N-[(1r)-3-(4-hydroxyphenyl)butanoyl]-2-decanamide (DB07991) as the best ligand, demonstrating favorable binding affinity and stability. Molecular dynamics simulations revealed strong protein-ligand interactions with consistent RMSD (~0.25 nm), RMSF (~0.5 nm), compact Rg (4.0-3.9 nm), and stable SASA profiles, indicating that the SLC4A4 structure remains stable upon ligand binding. CONCLUSIONS The findings suggest that DB07991 is a promising drug candidate for further investigation as a therapeutic agent against CRC, particularly for targeting SLC4A4. This study highlights the potential of computational drug repositioning in identifying effective treatments for colorectal cancer.
Collapse
Affiliation(s)
- Krunal Pawar
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur 303002, Rajasthan, India; (K.P.); (R.R.K.N.)
| | - Pramodkumar P. Gupta
- School of Biotechnology and Bioinformatics, D Y Patil Deemed to be University, Plot 50, Sector 15, CBD Belapur, Navi Mumbai 400614, Maharashtra, India
| | - Pooran Singh Solanki
- Bioinformatics Center, Birla Institute of Scientific Research, Jaipur 302001, Rajasthan, India;
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Off Campus Jaipur, Jaipur 302001, Rajasthan, India
| | - Ravi Ranjan Kumar Niraj
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur 303002, Rajasthan, India; (K.P.); (R.R.K.N.)
| | - Shanker L. Kothari
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur 303002, Rajasthan, India; (K.P.); (R.R.K.N.)
| |
Collapse
|
102
|
Liu H, Jin X, Liu S, Liu X, Pei X, Sun K, Li M, Wang P, Chang Y, Wang T, Wang B, Yu XA. Recent advances in self-targeting natural product-based nanomedicines. J Nanobiotechnology 2025; 23:31. [PMID: 39833846 PMCID: PMC11749302 DOI: 10.1186/s12951-025-03092-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
Natural products, recognized for their potential in disease prevention and treatment, have been integrated with advanced nano-delivery systems to create natural product-based nanomedicines, offering innovative approaches for various diseases. Natural products derived from traditional Chinese medicine have their own targeting effect and remarkable therapeutic effect on many diseases, but there are some shortcomings such as poor physical and chemical properties. The construction of nanomedicines using the active ingredients of natural products has become a key step in the modernization research process, which could be used to make up for the defects of natural products such as low solubility, large dosage, poor bioavailability and poor targeting. Nanotechnology enhances the safety, selectivity, and efficacy of natural products, positioning natural product-based nanomedicines as promising candidates in medicine. This review outlines the current status of development, the application in different diseases, and safety evaluation of natural product-based nanomedicines, providing essential insights for further exploration of the synergy between natural products and nano-delivery systems in disease treatment.
Collapse
Affiliation(s)
- Haifan Liu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xingyue Jin
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Suyi Liu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xinyue Liu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiao Pei
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Kunhui Sun
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Meifang Li
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Ping Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Yanxu Chang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tiejie Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Bing Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Xie-An Yu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
| |
Collapse
|
103
|
Lee PK, Co VA, Yang Y, Wan MLY, El-Nezami H, Zhao D. Bioavailability and interactions of schisandrin B with 5-fluorouracil in a xenograft mouse model of colorectal cancer. Food Chem 2025; 463:141371. [PMID: 39332376 DOI: 10.1016/j.foodchem.2024.141371] [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: 12/27/2023] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Schisandrin B (Sch B) is a predominant bioactive lignan from the fruit of a Chinese medicine food homology plant, Schisandra chinensis. Previously, we observed potent anti-tumor effect of Sch-B in colorectal cancer (CRC) and enhanced chemotherapy efficacy with fluorouracil (5-FU). However, their bioavailability and reciprocal interactions under CRC conditions are unclear. In this study, we first compared the bioavailability, metabolism and tissue distribution of Sch-B between non-tumor-bearing and xenograft CRC tumor-bearing mice. Next, we examined SchB-5-FU interactions via investigating alterations in drug metabolism and multidrug resistance. Using a validated targeted metabolomics approach, five active metabolites, including Sch-B and fluorodeoxyuridine triphosphate, were found tumor-accumulative. Co-treatment resulted in higher levels of Sch-B and 5-FU metabolites, showing improved phytochemical and drug bioavailability. Multidrug resistance gene (MDR1) was significantly downregulated upon co-treatment. Overall, we demonstrated the potential of Sch-B to serve as a promising chemotherapy adjuvant via improving drug bioavailability and metabolism, and attenuating MDR.
Collapse
Affiliation(s)
- Pui-Kei Lee
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, SAR, China; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong, SAR, China.
| | - Vanessa Anna Co
- School of Biological Sciences, The University of Hong Kong, Hong Kong, SAR, China; Department of Microbiology, The University of Hong Kong, and Centre for Virology, Vaccinology and Therapeutics, Hong Kong, SAR, China.
| | - Yang Yang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, SAR, China; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, SAR, China.
| | - Murphy Lam Yim Wan
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, United Kingdom; Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund 221 84, Sweden.
| | - Hani El-Nezami
- School of Biological Sciences, The University of Hong Kong, Hong Kong, SAR, China; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio FI-70211, Finland.
| | - Danyue Zhao
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, SAR, China; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong, SAR, China.
| |
Collapse
|
104
|
Haake LR, El Menuawy A, Rennau H, Marthe F, Hähnel U, Bock F, Hildebrandt G, Manda K. Viability and Radiosensitivity of Human Tumor Cells from Breast and Colon Are Influenced by Hypericum perforatum Extract HP01. Int J Mol Sci 2025; 26:622. [PMID: 39859336 PMCID: PMC11765585 DOI: 10.3390/ijms26020622] [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: 11/21/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
To enhance the treatment of tumors that are resistant to radio- and chemotherapy while minimizing the side effects of radiochemotherapy, researchers are continuously seeking new active compounds for use in combination with radiotherapy. Therefore, the aim of our study was to examine the cytotoxic and radiosensitizing effects of an extract from St. John's Wort (Hypericum perforatum), referred to as HP01, on human epithelial tumor cells in vitro. The growth of MCF-7 (breast carcinoma) and HT-29 (colon carcinoma) cells was examined under the influence of HP01. In combination with radiation, the effects of HP01 on cytotoxicity and long-term survival were assessed using a colony formation assay. The number of DNA double-strand breaks was analyzed using the γH2AX assay, while cell cycle distribution was examined via flow cytometry. A growth-inhibiting and cytotoxic effect was observed for both tumor cell lines starting at a concentration of 10 µg/mL HP01. Treatment with HP01 resulted in an inhibition of clonogenic survival of tumor cells after ionizing radiation (6 Gy). The number of DNA double-strand breaks (DSBs) in tumor cells increased with HP01 treatment, but the repair of radiation-induced DNA DSBs was not affected. Cell cycle analysis revealed that HP01, in addition to radiation, enhanced G2/M arrest in MCF-7 and HT-29 cells. Overall, HP01 not only showed a growth-inhibiting effect but also demonstrated a radiosensitizing effect on human tumor cells for the first time. We conclude that the HP01-induced G2/M accumulation of cells may be the main rationale for the drug-induced radiosensitivity. It is therefore a promising candidate for combined therapy in tumor diseases and warrants further investigation.
Collapse
Affiliation(s)
- Linda Rebecca Haake
- Department of Radiotherapy and Radiation Oncology, University Medical Center Rostock, Suedring 75, 18059 Rostock, Germany; (L.R.H.); (H.R.); (F.B.); (G.H.)
| | - Ahmed El Menuawy
- Institute for Breeding Research on Horticultural Crops, Julius Kühn Institute (JKI), Federal Research Centre for Cultivated Plants, Erwin-Baur-Strasse 27, 06484 Quedlinburg, Germany; (A.E.M.); (F.M.)
| | - Hannes Rennau
- Department of Radiotherapy and Radiation Oncology, University Medical Center Rostock, Suedring 75, 18059 Rostock, Germany; (L.R.H.); (H.R.); (F.B.); (G.H.)
| | - Frank Marthe
- Institute for Breeding Research on Horticultural Crops, Julius Kühn Institute (JKI), Federal Research Centre for Cultivated Plants, Erwin-Baur-Strasse 27, 06484 Quedlinburg, Germany; (A.E.M.); (F.M.)
| | - Urs Hähnel
- Institute for Breeding Research on Horticultural Crops, Julius Kühn Institute (JKI), Federal Research Centre for Cultivated Plants, Erwin-Baur-Strasse 27, 06484 Quedlinburg, Germany; (A.E.M.); (F.M.)
| | - Felix Bock
- Department of Radiotherapy and Radiation Oncology, University Medical Center Rostock, Suedring 75, 18059 Rostock, Germany; (L.R.H.); (H.R.); (F.B.); (G.H.)
| | - Guido Hildebrandt
- Department of Radiotherapy and Radiation Oncology, University Medical Center Rostock, Suedring 75, 18059 Rostock, Germany; (L.R.H.); (H.R.); (F.B.); (G.H.)
| | - Katrin Manda
- Department of Radiotherapy and Radiation Oncology, University Medical Center Rostock, Suedring 75, 18059 Rostock, Germany; (L.R.H.); (H.R.); (F.B.); (G.H.)
| |
Collapse
|
105
|
Li Y, Li C, Fan J, Liu Y, Xu Y, Pang G. The Application of Traditional Chinese Medicine-Derived Formulations in Cancer Immunotherapy: A Review. Cancer Manag Res 2025; 17:23-34. [PMID: 39816489 PMCID: PMC11734505 DOI: 10.2147/cmar.s503071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/21/2024] [Indexed: 01/18/2025] Open
Abstract
Background Cancer immunotherapy is an advanced therapeutic approach that harnesses the body's immune system to target and eliminate tumor cells. Traditional Chinese medicine (TCM), with a history rooted in centuries of clinical practice, plays a crucial role in enhancing immune responses, alleviating cancer-related symptoms, and reducing the risks of infections and complications in cancer patients. Methodology This review systematically examines the current literature on TCM-based formulations in cancer immunotherapy. It explores the mechanisms by which TCM augments immune responses, particularly focusing on how these formulations influence both innate and adaptive immunity. Various TCM herbs and compounds, their active ingredients, and their application in cancer prevention and treatment were analyzed through an integrated review of preclinical studies, clinical trials, and molecular mechanistic investigations. Results TCM formulations contribute to cancer therapy by modulating the body's internal environment to improve immune defense. They enhance the immune system's ability to detect and destroy cancer cells, promote apoptosis in tumor cells, inhibit tumor growth and metastasis, and augment the effectiveness of conventional cancer treatments. The review highlights specific TCM herbs and formulations that have demonstrated significant anti-cancer properties, including their ability to strengthen immune responses and provide synergistic effects with existing cancer therapies. Conclusion TCM-derived formulations represent a promising addition to cancer immunotherapy. The mechanisms through which these formulations enhance anti-tumor immunity are multifaceted, involving modulation of immune cell activity, apoptosis induction, and suppression of tumor progression. As cancer immunotherapy evolves, the integration of TCM into conventional treatment regimens may offer enhanced therapeutic efficacy, reduced side effects, and improved overall outcomes for cancer patients. Further clinical research is needed to fully elucidate the therapeutic potential and safety of TCM-based immunotherapeutic strategies in cancer care.
Collapse
Affiliation(s)
- Yanyun Li
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Changying Li
- School of Ophthalmology and optometry, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Junzi Fan
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yutong Liu
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yincong Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Guowei Pang
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
106
|
Cui Z, Liu X, E T, Lin H, Wang D, Liu Y, Ruan X, Wang P, Liu L, Xue Y. Effect of SNORD113-3/ADAR2 on glycolipid metabolism in glioblastoma via A-to-I editing of PHKA2. Cell Mol Biol Lett 2025; 30:5. [PMID: 39794701 PMCID: PMC11724473 DOI: 10.1186/s11658-024-00680-9] [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: 05/15/2024] [Accepted: 12/17/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive brain tumor, characterized by its poor prognosis. Glycolipid metabolism is strongly associated with GBM development and malignant behavior. However, the precise functions of snoRNAs and ADARs in glycolipid metabolism within GBM cells remain elusive. The objective of the present study is to delve into the underlying mechanisms through which snoRNAs and ADARs exert regulatory effects on glycolipid metabolism in GBM cells. METHODS RNA immunoprecipitation and RNA pull-down experiments were conducted to verify the homodimerization of ADAR2 by SNORD113-3, and Sanger sequencing and Western blot experiments were used to detect the A-to-I RNA editing of PHKA2 mRNA by ADAR2. Furthermore, the phosphorylation of EBF1 was measured by in vitro kinase assay. Finally, in vivo studies using nude mice confirmed that SNORD113-3 and ADAR2 overexpression, along with PHKA2 knockdown, could suppress the formation of subcutaneous xenograft tumors and improve the outcome of tumor-bearing nude mice. RESULTS We found that PHKA2 in GBM significantly promoted glycolipid metabolism, while SNORD113-3, ADAR2, and EBF1 significantly inhibited glycolipid metabolism. SNORD113-3 promotes ADAR2 protein expression by promoting ADAR2 homodimer formation. ADAR2 mediates the A-to-I RNA editing of PHKA2 mRNA. Mass spectrometry analysis and in vitro kinase testing revealed that PHKA2 phosphorylates EBF1 on Y256, reducing the stability and expression of EBF1. Furthermore, direct binding of EBF1 to PKM2 and ACLY promoters was observed, suggesting the inhibition of their expression by EBF1. These findings suggest the existence of a SNORD113-3/ADAR2/PHKA2/EBF1 pathway that collectively regulates the metabolism of glycolipid and the growth of GBM cells. Finally, in vivo studies using nude mice confirmed that knockdown of PHKA2, along with overexpression of SNORD113-3 and ADAR2, could obviously suppress GBM subcutaneous xenograft tumor formation and improve the outcome of those tumor-bearing nude mice. CONCLUSIONS Herein, we clarified the underlying mechanism involving the SNORD113-3/ADAR2/PHKA2/EBF1 pathway in the regulation of GBM cell growth and glycolipid metabolism. Our results provide a framework for the development of innovative therapeutic interventions to improve the prognosis of patients with GBM.
Collapse
Affiliation(s)
- Zheng Cui
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, Shenyang, China
- Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, Shenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Tiange E
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Hongda Lin
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Xuelei Ruan
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Ping Wang
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Libo Liu
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Yixue Xue
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China.
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
107
|
Li X, Huang H, Li Y, Feng Y, Wang J, Luo S, Chen Y, Zhang Y, Yan G, Nan L. Gualou Guizhi Granule inhibits microglia-mediated neuroinflammation to protect against neuronal apoptosis in vitro and in vivo. Front Immunol 2025; 15:1527986. [PMID: 39850889 PMCID: PMC11754197 DOI: 10.3389/fimmu.2024.1527986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Object Neuroinflammation mediated by microglia has emerged as a critical factor in ischemic stroke and neuronal damage. Gualou Guizhi Granule (GLGZG) has been shown to suppress inflammation in lipopolysaccharide (LPS)-activated microglia, though the underlying mechanisms and its protective effects against neuronal apoptosis remain unclear. This study aims to investigate how GLGZG regulates the Notch signaling pathway in microglia to reduce neuroinflammation and protect neurons from apoptosis. Method Using in vitro and in vivo models, we explored GLGZG's impact on microglia activation, pro-inflammatory cytokines, and neuronal apoptosis. Microglial cells were activated with LPS, and primary neuronal cells were exposed to LPS-activated microglia to simulate neuroinflammation. Additionally, we investigated the effects of GLGZG in combination with N-[N-[3,5-difluorophenacetyl]-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) or siRNA-Notch1 to further elucidate the involvement of the Notch signaling pathway. Results GLGZG significantly inhibited microglia activation and reduced neuroinflammation by de-creasing the levels of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α in both in vitro and in vivo models. GLGZG also effectively protected against microglia-induced neuronal apoptosis. Mechanistically, GLGZG down-regulated key components of the Notch signaling pathway, in-cluding Notch-1, NICD, RBPSUH, and Hes-1, in activated microglia. Combined treatment with GLGZG and DAPT or siRNA-Notch1 demonstrated enhanced inhibition of microglial activation and neuroinflammation. Conclusion Our findings reveal that GLGZG exerts its protective effects through the suppression of the Notch signaling pathway, thereby inhibiting microglia activation, reducing neuroinflammation, and safeguarding neurons from neuroinflammation-induced damage, offering potential as a therapeutic agent for ischemic stroke-induced neuroinflammation.
Collapse
Affiliation(s)
- Xuezhen Li
- Institute of Structural Pharmacology and Traditional Chinese Medicine (TCM) Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Han Huang
- Institute of Structural Pharmacology and Traditional Chinese Medicine (TCM) Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yanan Li
- Institute of Structural Pharmacology and Traditional Chinese Medicine (TCM) Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yi Feng
- Institute of Structural Pharmacology and Traditional Chinese Medicine (TCM) Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jinxuan Wang
- Pharmacy College, Fujian Medical University, Fuzhou, Fujian, China
| | - Shuping Luo
- Institute of Structural Pharmacology and Traditional Chinese Medicine (TCM) Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yaping Chen
- Institute of Structural Pharmacology and Traditional Chinese Medicine (TCM) Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yuqin Zhang
- Institute of Structural Pharmacology and Traditional Chinese Medicine (TCM) Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Guohong Yan
- Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Lihong Nan
- Institute of Structural Pharmacology and Traditional Chinese Medicine (TCM) Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
108
|
Lv Y, Hua Z, Lu X. Protective effects and possible mechanisms of mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles against kidney fibrosis in animal models: a systematic review and meta-analysis. Front Pharmacol 2025; 15:1511525. [PMID: 39830341 PMCID: PMC11739157 DOI: 10.3389/fphar.2024.1511525] [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: 10/15/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction The risk of kidney fibrosis is significantly elevated in individuals with diabetes, chronic nephritis, trauma, and other underlying conditions. Concurrently, human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) and their extracellular vesicles (MSC-Exos) have gained prominence in regenerative medicine. In light of these observations, we are undertaking a meta-analysis to elucidate the influence of hUCB-MSCs and MSC-Exos on kidney fibrosis. Methods To identify eligible trials, we conducted a comprehensive search of the CNKI, PubMed, Web of Science and Wanfang databases from inception to 24 October 2022. Furthermore, the methodological quality of the included studies was evaluated using the Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) risk-of-bias tool. Besides, the weighted standard mean difference (SMD) with a 95% confidence interval (CI) was calculated using the Review Manager 5.4 software. The Stata (12.0) software was employed to assess the impact of factors on outcome heterogeneity and publication bias in the study. A total of 645 related research studies were retrieved, of which 14 that involved 219 experimental animals were included in the study. Results In comparison to the control treatment, treatment with Human UCB MSC and MSC-Exos was observed to significantly enhance renal function in animal models of kidney fibrosis. This was evidenced by a reduction in serum creatinine (Scr) levels (p < 0.00001) and blood urea nitrogen (BUN) levels (p < 0.00001), as well as reduction of CD68+ macrophages (p < 0.00001), TdT-mediated dUTP Nick-End labeling (TUNEL)+ tubular cells(p < 0.00001), α-SMA levels (p = 0.0009) and TGF-β1 (p < 0.00001). P < 0.05 is deemed to indicate a statistically significant difference. Alpha-smooth muscle actin (α-SMA) is a specific protein that is normally expressed in myofibroblasts. The term "CD68+ macrophages" refers to macrophages that express the CD68 protein on their cell surface. Both macrophages and myofibroblasts have been linked to the development of kidney fibrosis. In this study, the quantity of CD68+ macrophages and α-SMA was employed as a means of gauging the extent of renal fibrosis. Additionally, transforming growth factor beta 1 (TGF-β1) is a significant cytokine implicated in the pathogenesis of kidney fibrosis. TUNEL-positive tubular cells represent tubular cells undergoing apoptosis. It is hypothesized that this may result in a reduction of tubular apoptosis and a delay in kidney fibrosis, due to the inhibition of the transformation of macrophages into myofibroblasts (MMT) and the disruption of the kidney fibrogenic niche. Conclusion The principal findings of this preclinical systematic review indicate that hUCB MSC and MSC-Exos have a substantial protective impact against kidney fibrosis. Kidney transfer remains the final option for traditional renal fibrosis treatment. The lack of donors and high cost make it challenging for many patients to access appropriate treatment. Although this study still suffers from three shortcomings: sample size, methodological consistency and translational challenges, the hUCB MSC and MSC-Exos have been demonstrated to reduce tubular apoptosis and inhibit fibrotic progression. The hUCB MSC and MSC-Exos offer a promising alternative due to their lower price and accessibility. Nevertheless, further high-quality studies are required in the future to address the methodological limitations identified in this review. Systematic Review Registration Identifier INPLASY2022100104.
Collapse
Affiliation(s)
- Yuanchen Lv
- 1st Clinical Department, China Medical University, Shenyang, Liaoning, China
| | - Zibo Hua
- 1st Clinical Department, China Medical University, Shenyang, Liaoning, China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
109
|
He X, Wang G, Wang Y, Zhang C. Matrine Enhances the Antitumor Efficacy of Chidamide in CTCL by Promoting Apoptosis. Recent Pat Anticancer Drug Discov 2025; 20:223-231. [PMID: 38571359 DOI: 10.2174/0115748928289036240318040756] [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: 10/24/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Cutaneous T-cell Lymphoma (CTCL) is a rare group of non-Hodgkin lymphoma originating from the skin, which is characterized by T-cell lymphoproliferative disorders. Chidamide, a Chinese original antineoplastic agent with independent intellectual property rights, and matrine, an extract of Chinese herbal medicine, both have been reported to exert effects on the treatment of tumors individually. However, chidamide combined with matrine has not been tested for the treatment of CTCL. METHODS Both HH and Hut78 CTCL cell lines were treated with chidamide (0.4 μmol/L), matrine (0.6 g/L), or chidamide combined with matrine for 24, 48, and 72 h. Cell viability was estimated by MTS assay at each time point. Flow cytometry was then conducted to detect cell apoptosis. The exact mechanism of chidamide combined with matrine on CTCL cells was detected by Western blotting and further validated in xenograft models of NOD/SCID mice. RESULTS AND DISCUSSION Compared to the single drug, chidamide combined with matrine showed a more significant effect on proliferation inhibition and apoptosis induction on CTCL cells both in vitro and in vivo. The results from the in vitro and in vivo studies suggested that matrine could enhance the anti-tumor effect of chidamide by increasing the protein expression of cleaved caspase- 3 and decreasing the expression of E-cadherin, NF-κB, p-Bad, and Bcl-2 to activate apoptosis. CONCLUSION Our data have demonstrated chidamide combined with matrine to exhibit elevated antitumor activity in both CTCL cells and xenograft models of NOD/SCID mice, which may be a potential treatment option for CTCL.
Collapse
Affiliation(s)
- Xinglan He
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Guanyu Wang
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Yimeng Wang
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Chunlei Zhang
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
110
|
Sun C, Zeng B, Zhou J, Li N, Li M, Zhu C, Xie S, Wang Y, Wang S, Wang X. Analysis of SLC genes alternative splicing identifies the SLC7A6 RI isoform as a therapeutic target for colorectal cancer. Cancer Sci 2025; 116:233-247. [PMID: 39403788 PMCID: PMC11711054 DOI: 10.1111/cas.16351] [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: 07/03/2024] [Revised: 08/30/2024] [Accepted: 09/07/2024] [Indexed: 01/11/2025] Open
Abstract
Alternative splicing (AS), a crucial mechanism in post-transcriptional regulation, has been implicated in diverse cancer processes. Several splicing variants of solute carrier (SLC) transporters reportedly play pivotal roles in tumorigenesis and tumor development. However, an in-depth analysis of AS landscapes of SLCs in colon adenocarcinoma (COAD) is lacking. Herein, we analyzed data from The Cancer Genome Atlas and identified 1215 AS events across 243 SLC genes, including 109 differentially expressed AS (DEAS) events involving 62 SLC genes in COAD. Differentially spliced SLCs were enriched in biological processes, including transmembrane transporter activity, transporter activity, ferroptosis, and choline metabolism. In patients with COAD, tumor tissues exhibited higher expression of longer mitochondrial carrier SLC25A16 isoforms than adjacent normal tissues, consistent with bioinformatics analysis. Protein-coding sequences and transmembrane helices of survival-related DEAS were predicted, revealing that shifts in splicing sites altered the number and structure of their transmembrane proteins. We developed a prognostic risk model based on the screened 6-SLC-AS (SLC7A6_RI_37208 (SLC7A6-RI), SLC11A2_AP_21724, SLC2A8_ES_87631, SLC35B1_AA_42317, SLC39A11_AD_43204, and SLC7A8_AP_26712). Knockdown of the intronic region of SLC7A6-RI isoform enhanced colon cancer cell proliferation. In vivo, knockdown of the intronic region of SLC7A6-RI isoform enhanced tumor growth in colon cancer. Mechanistically, si-SLC7A6-RI isoform exerted oncogenic effects by activating the PI3K-Akt-mTOR signaling pathway and promoting cell proliferation, evidenced by increased expression of key regulators Phosphorylated Mammalian Target of Rapamycin (p-mTOR) and a cell proliferation marker Proliferating Cell Nuclear Antigen (PCNA) using western blotting. Our study elucidated SLC-AS in COAD, highlighting its potential as a prognostic and therapeutic target and emphasizing the suppressive influence of SLC7A6-RI in colon cancer progression.
Collapse
Affiliation(s)
- Chao Sun
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
- Integrated Chinese and Western Medicine Postdoctoral Research StationJinan UniversityGuangzhouChina
| | - Boning Zeng
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
- Integrated Chinese and Western Medicine Postdoctoral Research StationJinan UniversityGuangzhouChina
| | - Jilong Zhou
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Nan Li
- School of Pharmaceutical SciencesShenzhen University Health Science CenterShenzhenChina
| | - Mingwei Li
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Chaowei Zhu
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Shouxia Xie
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| | - Yifei Wang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw MaterialsJinan UniversityGuangzhouChina
| | - Shaoxiang Wang
- School of Pharmaceutical SciencesShenzhen University Health Science CenterShenzhenChina
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's HospitalThe Second Clinical Medical College, Jinan UniversityShenzhenChina
| |
Collapse
|
111
|
Xin Y, Zhou S, Chu T, Zhou Y, Xu A. Protective Role of Electroacupuncture Against Cognitive Impairment in Neurological Diseases. Curr Neuropharmacol 2025; 23:145-171. [PMID: 38379403 PMCID: PMC11793074 DOI: 10.2174/1570159x22999240209102116] [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: 08/12/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 02/22/2024] Open
Abstract
Many neurological diseases can lead to cognitive impairment in patients, which includes dementia and mild cognitive impairment and thus create a heavy burden both to their families and public health. Due to the limited effectiveness of medications in treating cognitive impairment, it is imperative to develop alternative treatments. Electroacupuncture (EA), a required method for Traditional Chinese Medicine, has the potential treatment of cognitive impairment. However, the molecular mechanisms involved have not been fully elucidated. Considering the current research status, preclinical literature published within the ten years until October 2022 was systematically searched through PubMed, Web of Science, MEDLINE, Ovid, and Embase. By reading the titles and abstracts, a total of 56 studies were initially included. It is concluded that EA can effectively ameliorate cognitive impairment in preclinical research of neurological diseases and induce potentially beneficial changes in molecular pathways, including Alzheimer's disease, vascular cognitive impairment, chronic pain, and Parkinson's disease. Moreover, EA exerts beneficial effects through the same or diverse mechanisms for different disease types, including but not limited to neuroinflammation, neuronal apoptosis, neurogenesis, synaptic plasticity, and autophagy. However, these findings raise further questions that need to be elucidated. Overall, EA therapy for cognitive impairment is an area with great promise, even though more research regarding its detailed mechanisms is warranted.
Collapse
Affiliation(s)
- Yueyang Xin
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tiantian Chu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaqun Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aijun Xu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
112
|
Vazhappilly CG, Alsawaf S, Mathew S, Nasar NA, Hussain MI, Cherkaoui NM, Ayyub M, Alsaid SY, Thomas JG, Cyril AC, Ramadan WS, Chelakkot AL. Pharmacodynamics and safety in relation to dose and response of plant flavonoids in treatment of cancers. Inflammopharmacology 2025; 33:11-47. [PMID: 39580755 DOI: 10.1007/s10787-024-01581-1] [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: 08/08/2024] [Accepted: 10/02/2024] [Indexed: 11/26/2024]
Abstract
Despite the recent advancements in developing bioactive nutraceuticals as anticancer modalities, their pharmacodynamics, safety profiles, and tolerability remain elusive, limiting their success in clinical trials. The failure of anticancer drugs in clinical trials can be attributed to the changes in drug clearance, absorption, and cellular responses, which alter the dose-response efficacy, causing adverse health effects. Flavonoids demonstrate a biphasic dose-response phenomenon exerting a stimulatory or inhibitory effect and often follow a U-shaped curve in different preclinical cancer models. A double-edged sword, bioflavonoids' antioxidant or prooxidant properties contribute to their hormetic behavior and facilitate redox homeostasis by regulating the levels of reactive oxygen species (ROS) in cells. Emerging reports suggest a need to discuss the pharmacodynamic broad-spectrum of plant flavonoids to improve their therapeutic efficacy, primarily to determine the ideal dose for treating cancer. This review discusses the dose-response effects of a few common plant flavonoids against some types of cancers and assesses their safety and tolerability when administered to patients. Moreover, we have emphasized the role of dietary-rich plant flavonoids as nutraceuticals in cancer treatment and prevention.
Collapse
Affiliation(s)
- Cijo George Vazhappilly
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE.
| | - Seba Alsawaf
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Shimy Mathew
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, UAE
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah, UAE
| | - Noora Ali Nasar
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Maheen Imtiaz Hussain
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Noor Mustapha Cherkaoui
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Mohammed Ayyub
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Serin Yaser Alsaid
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Joshua George Thomas
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
| | - Asha Caroline Cyril
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, UAE
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Wafaa S Ramadan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
| | | |
Collapse
|
113
|
Chen J, Zhang X. An Integrated Approach of Network Pharmacology, Bioinformatics, Molecular Docking, and Experimental Verification Uncovers Prunellae Spica as the Potential Medicine of Prognosis Improvement for Oral Squamous Cell Carcinoma. Curr Pharm Des 2025; 31:391-412. [PMID: 39289945 DOI: 10.2174/0113816128328547240827045955] [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: 06/07/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Prunellae Spica (PS), the spike from Prunella vulgaris L., is a traditional Chinese medicine that can treat Oral Squamous Cell Carcinoma (OSCC), whereas its molecular mechanisms and effects on the prognosis of patients remain unclear. METHODS Our study aimed to identify the potential anti-OSCC targets of PS and explore its mechanisms and effects on prognosis through network pharmacology, bioinformatics analysis, molecular docking, and in-vitro cell assays. RESULTS Sixty-two potential targets of 11 active anti-OSCC ingredients of PS were identified, with Quercetin, the core ingredient of PS, exhibiting the most significant number of OSCC-related targets. GO analysis indicated that the primary biological processes involved in OSCC treatment by PS were the cellular response to nitrogen compound, response to xenobiotic stimulus, and cellular response to organonitrogen compound. KEGG analysis revealed that pathways in cancer were the top highly enriched signaling pathway in the treatment of OSCC by PS. DisGeNET analysis is mainly about Lip and Oral Cavity Carcinoma. More importantly, 6 of the 62 targets were markedly related to prognosis. Molecular docking revealed high affinities between the key component and the prognosis-related target proteins. Treatment of OSCC cell line SCC-25 with Quercetin could inhibit malignant biological behaviors, such as cell proliferation, colony formation, invasion, and migration, as well as affect the targets related to prognosis and promote autophagy. CONCLUSION Overall, these results suggest that PS plays a significant role in treating and improving the prognosis of OSCC by directly influencing various processes in OSCC.
Collapse
MESH Headings
- Humans
- Molecular Docking Simulation
- Network Pharmacology
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/pathology
- Mouth Neoplasms/diagnosis
- Mouth Neoplasms/metabolism
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/diagnosis
- Carcinoma, Squamous Cell/metabolism
- Computational Biology
- Prunella/chemistry
- Cell Proliferation/drug effects
- Prognosis
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/isolation & purification
- Drug Screening Assays, Antitumor
Collapse
Affiliation(s)
- Jiahui Chen
- Beijing Stomatological Hospital and School of Stomatology, Beijing Institute of Dental Research, Capital Medical University, No. 4 Tiantanxili, Dongcheng District, Beijing 100050, China
| | - Xinyan Zhang
- Beijing Stomatological Hospital and School of Stomatology, Beijing Institute of Dental Research, Capital Medical University, No. 4 Tiantanxili, Dongcheng District, Beijing 100050, China
| |
Collapse
|
114
|
Wang Q, Li S, Fan Y, Chen W, Jiang Q, Sun X, Lv Q, Li W, Jia Y. Yiqi Jianpi Kangai Decoction Enhances the Chemotherapy Effect by Inducing Apoptosis and Regulating Treg and Th17 Cells in Colorectal Cancer Mice Model with Spleen Qi Deficiency. J Evid Based Integr Med 2025; 30:2515690X241313097. [PMID: 39980387 PMCID: PMC11843709 DOI: 10.1177/2515690x241313097] [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: 03/21/2023] [Revised: 06/10/2024] [Accepted: 12/16/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Colorectal Cancer (CRC) is widely prevalent worldwide and its incidence is increasing. Chemotherapy is an important treatment method for colorectal cancer in addition to surgery, but it often causes physical and mental pain to patients due to its side effects. TCM emphasizes evidence-based treatment and a holistic concept, and the combination of TCM and chemotherapy can reduce chemotherapy side effects, improve chemotherapy efficacy, and enhance patients' immunity. Yiqi Jianpi Kangai Decoction (YQJP) has been used clinically to treat patients with advanced colorectal cancer and may improve their immune function and prognosis. However, its mechanism has not been elucidated. OBJECTIVE The study aims to explore the effect and mechanism of YQJP on enhancing the therapeutic effect of chemotherapy on spleen qi deficiency type CRC mice. METHODS We used HPLC/MS to characterize the active substance components in YQJP. We established the spleen qi deficiency model induced by using the compound methods of diarrhea of bitter and cold, starvation, and full and excessive labor. and then inoculated CT-26 mouse colon cancer cells subcutaneously to establish the CRC Mice. We also evaluated the efficacy and mechanism of YQJP by using oral Yiqi Jianpi Kangai Decoction combined with an intraperitoneal injection of chemotherapeutic drugs (FOLFOX regimen). We evaluated the efficacy and safety of YQJP by observing the general condition (body weight, tumor size, food intake, hair condition, stool condition), HE staining, blood routine, and organ index of the mice. The expression of CD8+, CD4+ T cells, Th17 cells, and Treg cells in the tumor and spleen were detected by flow cytometry, and the levels of IL-17, IL-10, IFN-γ and TGF-β in the tumor were detected by ELISA; the expression of Ki-67, PCNA, RORγt and FOXP3 proteins in the tumor was detected by immunohistochemistry. RESULTS YQJP contains 7 herbal compounds, which can effectively improve body weight, spleen condition, and bone marrow suppression in tumor-bearing mice inhibit tumor growth, and do not damage tissues and organs, which initially confirmed the anti-cancer effect and safety of YQJP. Further experiments showed that YQJP could elevate the proportion of CD8+, CD4+ T cells in the spleen, increase the proportion of Th17 cells in the tumor tissue of mice, and decrease the level of Treg cells. It can inhibit the expression of Ki-67 and PCNA. Meanwhile, it promotes the expression of IL-17 and IFN-γ and inhibits the expression of IL-10 and TGF-β. In addition, it can reduce the relative expression of FOXP3 and increase the relative expression of RORγt. CONCLUSION The combination of YQJP with chemotherapy had the effect of tumor suppression and enhanced chemotherapeutic efficacy in the spleen qi deficiency CRC mice. The related mechanism may be related to inhibiting proliferation, promoting apoptosis of tumor cells, increasing Th17 cells in tumor tissues, and decreasing Treg cell expression to improve the tumor microenvironment.
Collapse
Affiliation(s)
- Qinsha Wang
- The First Clinical Medical School of Guizhou University of Chinese Medicine, Guiyang, Guizhou Province, China
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Shichao Li
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yao Fan
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Weidong Chen
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qingfeng Jiang
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xin Sun
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qijun Lv
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Wusheng Li
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yingtian Jia
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
115
|
Zheng Q, Wu X, Peng S. The immunotherapy mechanism of Hedyotis Diffusae Herba in treating liver cancer: a study based on network pharmacology, bioinformatics, and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:951-965. [PMID: 39093467 DOI: 10.1007/s00210-024-03312-3] [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: 06/16/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
Liver cancer is a malignant tumor that develops on or inside the liver. Hedyotis diffusa Willd (HDW) plays a significant role in anti-tumor activities; however, its mechanism against liver cancer remains unclear. This study aims to evaluate the immunotherapeutic mechanism of HDW in treating liver cancer through network pharmacology, bioinformatics analysis, and experimental validation. Network pharmacology was utilized to identify the active components and potential targets of HDW from the TCMSP database. A potential target protein-protein interaction (PPI) network was constructed using the STRING database, followed by function and pathway enrichment analysis of the targets using GO and KEGG methods. In addition, the key targets for HDW against liver cancer were identified using five different algorithms in Cytoscape. The TCGA and HPA databases were used to assess the mRNA and protein expression of core target genes in normal liver and liver cancer tissues and their relationship with overall survival in liver cancer, as well as their role in immune infiltration. Molecular docking between the core components of HDW and the core targets was performed using PyMOL software. The effects of HDW on the proliferation and apoptosis of liver cancer cells were examined using MTT and flow cytometry. The regulatory effects of the core component quercetin on core targets were validated using RT-qPCR and Western blot. A total of 163 potential targets were identified by searching for intersections among 7 types of active components and all potential and liver cancer targets. PPI network analysis revealed the core targets IL6 and TNF. GO enrichment analysis involved 2089 biological processes, 76 cellular components, and 196 molecular functions. KEGG enrichment analysis suggested that the anti-cancer effects of HDW might be mediated by the AGE-RAGE signaling pathway, IL-17 signaling pathway, TNF signaling pathway, PI3K-Akt signaling pathway, and NF-κB signaling pathway. Database validation of key targets showed that mRNA and protein expression results for the IL6 gene were contradictory, while those for the TNF gene were consistent, both being underexpressed in liver cancer. Importantly, the expression of IL6 and TNF was related to the infiltration of 24 types of immune cells, with the highest correlation with macrophages. Molecular docking showed that IL6 and TNF had high binding stability with quercetin, with binding energies of - 7.4 and - 6.0 kJ∙mol-1, respectively. Experimental validation showed that quercetin inhibited liver cancer cell proliferation and promoted apoptosis in a dose-dependent manner, with protein results indicating that quercetin downregulated the mRNA and protein expression of IL6 and TNF, and upregulated key proteins in the AGE-RAGE signaling pathway, AGEs, and RAGE. This study comprehensively elucidates the activity, potential targets, and molecular mechanisms of HDW against liver cancer, providing a promising strategy for the scientific basis and treatment mechanism of traditional Chinese medicine in treating liver cancer.
Collapse
Affiliation(s)
- Qingsheng Zheng
- Department of General Surgery, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, 602 Ba Yi Qi Zhong Road, Taijiang District, Fuzhou, 350108, Fujian, China
| | - Xueying Wu
- Department of General Surgery, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, 602 Ba Yi Qi Zhong Road, Taijiang District, Fuzhou, 350108, Fujian, China
| | - Shuai Peng
- Department of General Surgery, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, 602 Ba Yi Qi Zhong Road, Taijiang District, Fuzhou, 350108, Fujian, China.
| |
Collapse
|
116
|
Huo M, Gao Z, Wang G, Hou Z, Zheng J. Huaier promotes sensitivity of colorectal cancer to oxaliplatin by inhibiting METTL3 to regulate the Wnt/β‑catenin signaling pathway. Oncol Rep 2025; 53:7. [PMID: 39513580 PMCID: PMC11574703 DOI: 10.3892/or.2024.8840] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Colorectal cancer (CRC) ranks fifth in terms of incidence rate and mortality among malignant tumors in China. Oxaliplatin (OXA) is a first‑line drug for the clinical treatment of CRC, but its antitumor effect is limited because of the development of drug resistance. The present study aimed to investigate whether the traditional Chinese medicine Huaier can regulate the Wnt/β‑catenin signaling pathway by affecting the expression of METTL3, thereby promoting the sensitivity of HCT‑8/L cells to OXA. The expression of METTL3 was analyzed based on the UCSC Xena and Gene Expression Omnibus databases. Silent METTL3 and overexpression METTL3 models were constructed, and Cell Counting Kit‑8 and flow cytometry were used to detect the effects of Huaier on the viability and apoptosis of HCT‑8/L cells. Western blotting, reverse transcription‑quantitative PCR, nuclear cytoplasmic separation and immunofluorescence were used to detect the effects of Huaier on the expression of METTL3, Pgp, Wnt/β‑catenin signaling pathway‑related proteins, apoptosis‑related proteins and related mRNA. The results demonstrated that patients with high expression levels of METTL3 had a shorter overall survival period. The expression level of METTL3 significantly increased in drug‑resistant CRC cells. Silencing METTL3 promoted apoptosis of CRC cells and increased their sensitivity to OXA by inhibiting the Wnt/β‑catenin signaling pathway. Huaier downregulated the expression of METTL3, thereby promoting apoptosis of drug‑resistant CRC cells and increasing their sensitivity to OXA by inhibiting the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Mingyi Huo
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Zhixu Gao
- Department of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Guizhen Wang
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Zhiping Hou
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Jining Zheng
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
117
|
Du W, Tang Z, Du A, Yang Q, Xu R. Bidirectional crosstalk between the epithelial-mesenchymal transition and immunotherapy: A bibliometric study. Hum Vaccin Immunother 2024; 20:2328403. [PMID: 38502119 PMCID: PMC10956627 DOI: 10.1080/21645515.2024.2328403] [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: 12/19/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
Immunotherapy has recently attracted considerable attention. However, currently, a thorough analysis of the trends associated with the epithelial-mesenchymal transition (EMT) and immunotherapy is lacking. In this study, we used bibliometric tools to provide a comprehensive overview of the progress in EMT-immunotherapy research. A total of 1,302 articles related to EMT and immunotherapy were retrieved from the Web of Science Core Collection (WOSCC). The analysis indicated that in terms of the volume of research, China was the most productive country (49.07%, 639), followed by the United States (16.89%, 220) and Italy (3.6%, 47). The United States was the most influential country according to the frequency of citations and citation burstiness. The results also suggested that Frontiers in Immunotherapy can be considered as the most influential journal with respect to the number of articles and impact factors. "Immune infiltration," "bioinformatics analysis," "traditional Chinese medicine," "gene signature," and "ferroptosis" were found to be emerging keywords in EMT-immunotherapy research. These findings point to potential new directions that can deepen our understanding of the mechanisms underlying the combined effects of immunotherapy and EMT and help develop strategies for improving immunotherapy.
Collapse
Affiliation(s)
- Wei Du
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| | - Zemin Tang
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| | - Ashuai Du
- Department of Infectious Diseases, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Qinglong Yang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
- Department of General Surgery, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Rong Xu
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| |
Collapse
|
118
|
Li Y, Guo C, Zhang F, Cheng S, Li Y, Luo S, Zeng Y, Zhao Y, Wu K. DNMT1 inhibition improves the activity of memory-like natural killer cells by enhancing the level of autophagy. Mol Biol Rep 2024; 52:68. [PMID: 39704855 DOI: 10.1007/s11033-024-10181-9] [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: 10/09/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a common hematological tumor, but it is difficult to treat. DNMT1 is a DNA methyltransferase whose main function is to maintain stable DNA methylation during the DNA replication process. DNMT1 also plays an important role in AML, but its function in cytokine-induced memory-like natural killer (CIML NK) cell activity remains unclear. METHODS AND RESULTS In this study, we isolated primary NK cells from the peripheral blood of healthy volunteers and AML patients and treated them with 10 ng/mL IL-12, 50 ng/mL IL-15 and 50 ng/mL IL-18 to promote their differentiation into CIML NK cells. The activity of CIML NK cells was evaluated by RT‒qPCR, western blotting, ELISAs, and flow cytometry. DNMT1 was highly expressed in NK cells from AML patients. Knocking down DNMT1 significantly increased the expression of CD25, CD137, CD107a, IFN-γ, and TNF-α and increased the activity of CIML NK cells. Mechanistically, knocking down DNMT1 promoted autophagy by activating the AMPK/mTOR signaling pathway, thereby enhancing the activity of CIML NK cells and alleviating the progression of AML. CONCLUSIONS Our study revealed that the downregulation of DNMT expression may be a new target for the treatment of AML.
Collapse
Affiliation(s)
- Yixun Li
- Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Chong Guo
- Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Fujia Zhang
- Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Shenju Cheng
- Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Yanhong Li
- Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Shan Luo
- Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Yun Zeng
- Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Yaling Zhao
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China.
| | - Kun Wu
- Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China.
| |
Collapse
|
119
|
Huang Z, Li F, Zheng X, Zheng J, Dong Y, Ding Z, Gou H, Yao M, Liu J. Catalpol promotes hippocampal neurogenesis and synaptogenesis in rats after multiple cerebral infarctions by mitochondrial regulation: involvement of the Shh signaling pathway. Front Pharmacol 2024; 15:1461279. [PMID: 39749196 PMCID: PMC11693731 DOI: 10.3389/fphar.2024.1461279] [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: 07/08/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction Ischemic stroke greatly threatens human life and health. Neuro-restoration is considered to be the critical points in reestablishing neurological function and improving the quality of life of patients. Catalpol is the main active ingredient of the Chinese herbal medicine Dihuang, which has the beneficial efficacy in traditional remedy, is closely related to the mitochondrial morphology and function. In the present study, we investigated whether catalpol has a neurorestorative effect after multiple cerebral infarctions and its underlying mechanisms. Methods In this study, male 8-week-old Sprague-Dawley (SD) rats were grouped according to neurological deficit scores to minimize differences between groups the second day: sham group, model group, Ginkgo biloba P.E (EGb) (Ginaton:18 mg/kg) group, model + CAT 30 mg/kg group (CAT 30), model + CAT 60 mg/kg group (CAT 60), and model + CAT 120 mg/kg group (CAT 120). From the first day to the fourteenth day after MCI, rats were given the corresponding doses of drugs by gastric administration every day(1 mL/100g), and from day 7 to day 14, all rats were injected with Brdu solution (50 mg/kg) i.p. Neuro-Function was assessed by the neurologic deficit scores. Then we observed measurement of brain atrophy and fluorescent Nissl staining. The expression of BrdU+/DCX+ cells and the BDNF concentrations were tested to observe the neuro-restoration effect. Transmission electron microscope (TEM) and Western blot (WB) were used to observed synaptogenesis. we observed the restoration of mitochondrial function by detecting the intracortical calcium and T-AOC content. Finally, we examined the protein and mRNA expression of shh signaling pathway through q-PCR and WB. Results Catalpol alleviated neurological deficits, reduced the degree of brain atrophy, as well as minimize pathological damage in the hippocampus and cortex. In addition, catalpol also promoted hippocampal neurogenesis and synaptogenesis by improving the mitochondrial structure and promoting mitochondrial function, as evidenced by the up-regulation of positive expression of both Recombinant Doublecortin (DCX) and 5-Bromodeoxyuridinc (BrdU), the enhancement of the Total antioxidant capacity (T-AOC), and the increase in the expression of synapse-associated proteins, Synaptophysin (SYP) and post-synaptic density-95 (PSD-95). Finally, we observed that catalpol up-regulated the expression of Sonic hedgehog (Shh) and Glioma-associated homologue-1 (GLI-1), factors related to the Shh signaling pathway. Discussion In conclusion, catalpol may regulate mitochondria through activation of the Shh signaling pathway and exert its role in promoting hippocampal neurogenesis and synaptogenesis.
Collapse
Affiliation(s)
- Zishan Huang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Feng Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Xiaoyu Zheng
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Jiarui Zheng
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Heilongjiang Academy of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yilei Dong
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Zhao Ding
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Huanyu Gou
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Mingjiang Yao
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Jianxun Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
120
|
Sui X, Gao B, Zhang L, Wang Y, Ma J, Wu X, Zhou C, Liu M, Zhang L. Scutellaria barbata D.Don and Hedyotis diffusa Willd herb pair combined with cisplatin synergistically inhibits ovarian cancer progression through modulating oxidative stress via NRF2-FTH1 autophagic degradation pathway. J Ovarian Res 2024; 17:246. [PMID: 39702302 DOI: 10.1186/s13048-024-01570-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Cisplatin (DDP) is one of the most effective anticancer drugs, commonly used to treat advanced ovarian cancer (OC). However, DDP has significant limitations of platinum-based drugs, including chemical resistance and high-dose toxic side effects. Traditional Chinese medicines (TCMs) often presented in the form of formula, in which the herb pair was the basic unit. Scutellaria barbata D.Don and Hedyotis diffusa Willd (SB-HD) are famous TCMs herb pair that have been shown to help treat multiple types of cancers. However, the synergistic effects and mechanism of combination of SB-HD and DDP to enhance DDP chemosensitivity in OC are still unknown. RESULTS In vitro, we found that the optimal proportion of SB-HD to inhibit the proliferation of OC cells was 2:1, SB-HD and DDP were shown to synergistically reduce the viability of OC cells, inhibit the colony formation, promote cell cycle arrest and apoptosis, as well as inhibit cell migration and invasion. In vivo, combination treatment significantly inhibited the growth of subcutaneous tumors in BALB/c nude mice and reduced the toxic side effects of DDP. Mechanistically, SB-HD and DDP combination treatment significantly promoted oxidative stress response, decreased MMP, inhibited ATP production, decreased ROS levels and increased SOD activity, increased the expression of NRF2, HO-1, ATG5 and LC3, decreased the expression of p62 and FTH1 both in OC cells and tumor tissue of mice. Inhibitor 3-MA (Methyladenine, autophagy inhibitor) and Fer-1 (Ferrostatin-1, iron ion inhibitor) can effectively reverse the expression changes of the key target proteins, but not ZnPP (Zinc protoporphyrin, HO-1 inhibitor). Through bioinformatics analysis, it was found that the abnormal expression level of NRF2 and FTH1 mRNA has a high prognostic value, at the same time, the other four key proteins respectively or interacting with NRF2 and FTH1, also play important roles in the occurrence and development of OC. CONCLUSION Our findings uncover a synergistic effect of SB-HD and DDP against OC through modulating oxidative stress via NRF2-FTH1 autophagic degradation pathway, which may provide an important theoretical foundation for the use of SB-HD and a new strategy for enhancing DDP chemosensitivity as well as reducing toxic side effects.
Collapse
Affiliation(s)
- Xue Sui
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Bingqing Gao
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- School of Pharmacy, Anhui Xinhua University, Hefei, 230088, China
| | - Liu Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- Department of Dermatology, Dalian Lvshunkou District Hospital of Traditional Chinese Medicine, Dalian, 116041, China
| | - Yanmin Wang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Junnan Ma
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xingchen Wu
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Chenyu Zhou
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Min Liu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| | - Lin Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
121
|
Liu ZQ, Yan CZ, Zhong SM, Chong CJ, Wu YQ, Liu JY, Huang CX, Wang KY, Li HW, Song JL. Dietary Antrodia cinnamomea Polysaccharide Intervention Modulates Clinical Symptoms by Regulating Ovarian Metabolites and Restructuring the Intestinal Microbiota in Rats with Letrozole-Induced PCOS. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27884-27901. [PMID: 39632724 DOI: 10.1021/acs.jafc.4c06855] [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: 12/07/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder. This study investigated the mitigating effects of the Antrodia cinnamomea polysaccharide (ACP) on a letrozole-induced PCOS rat model. Results demonstrated that ACP reduced obesity and ameliorated dyslipidemia in PCOS rats. Moreover, ACP restored estrous cycle regularity, suppressed polycystic ovarian changes, and regulated serum levels of sex hormones, SOD, and MDA. Furthermore, ACP increased the α-diversity and modulated the abundance of phyla (Bacteroidetes, Firmicutes, and Verrucomicrobia) and genera (Lactobacillus, Helicobacter, Akkermansia, Oscillospira, Coprococcus, Roseburia, Blautia, and Allobaculum) in the gut microbiota. ACP also restored compromised intestinal barriers by upregulating the expression of ZO1, Occludin, Claudin1, and Claudin7 in the colon. ACP mitigated ovarian fibrosis by preventing activation of the NLRP3 inflammasome, decreasing the expression of fibrotic markers (TGF-β1, collagen-I, α-SMA, and CTGF), and regulating four ovarian fibrosis-associated metabolomics pathways. Generally, dietary ACP effectively ameliorated clinical symptoms and inhibited ovarian fibrosis in PCOS rats.
Collapse
Affiliation(s)
- Zhi-Qiang Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guilin Medical University, Guilin 541004, China
| | - Chuan-Zhi Yan
- Department of Pharmacology, School of Pharmaceutical Sciences, Guilin Medical University, Guilin 541004, China
| | - Shu-Mei Zhong
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, Guangxi 541100, China
| | - Chao-Jie Chong
- Department of Pharmacology, School of Pharmaceutical Sciences, Guilin Medical University, Guilin 541004, China
| | - Ya-Qi Wu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guilin Medical University, Guilin 541004, China
| | - Jun-Yang Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, Guangxi 541100, China
| | - Chun-Xiang Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, Guangxi 541100, China
| | - Ke-Ying Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, Guangxi 541100, China
| | - He-Wei Li
- Department of Infectious Diseases, School of Clinical Medicine, Guilin Medical University, Guilin 541004, China
| | - Jia-Le Song
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, Guangxi 541100, China
- Guangxi Key Laboratory of Environmental Exposureomics and Entire Lifecycle Health, Guilin Medical University, Guilin, Guangxi 541100, China
- Department of Clinical Nutrition and Obstetrics, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, China
| |
Collapse
|
122
|
Shodry S, Hasan YTN, Ahdi IR, Ulhaq ZS. Gene targets with therapeutic potential in hepatocellular carcinoma. World J Gastrointest Oncol 2024; 16:4543-4547. [PMID: 39678796 PMCID: PMC11577361 DOI: 10.4251/wjgo.v16.i12.4543] [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: 06/27/2024] [Revised: 08/03/2024] [Accepted: 08/13/2024] [Indexed: 11/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide. Major treatments include liver transplantation, resection, and chemotherapy, but the 5-year recurrence rate remains high. Late diagnosis often prevents surgical intervention, contributing to poor patient survival rates. Carcinogenesis in HCC involves genetic alterations that drive the transformation of normal cells into malignant ones. Enhancer of zeste homolog 2 (EZH2), a key regulator of cell cycle progression, is frequently upregulated in HCC and is associated with advanced stages and poor prognosis, making it a potential biomarker. Additionally, signal transducer and activator of transcription 3, which binds to EZH2, affects disease staging and outcomes. Targeting EZH2 presents a promising therapeutic strategy. On the other hand, abnormal lipid metabolism is a hallmark of HCC and impacts prognosis. Fatty acid binding protein 5 is highly expressed in HCC tissues and correlates with key oncogenes, suggesting its potential as a biomarker. Other genes such as guanine monophosphate synthase, cell division cycle associated 5, and epidermal growth factor receptor provide insights into the molecular mechanisms of HCC, offering potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Syifaus Shodry
- Faculty of Medicine and Health Sciences, Maulana Ibrahim Islamic State University of Malang, Malang 65144, Jawa Timur, Indonesia
| | - Yuliono Trika Nur Hasan
- Faculty of Medicine and Health Sciences, Maulana Ibrahim Islamic State University of Malang, Malang 65144, Jawa Timur, Indonesia
| | - Iwal Reza Ahdi
- Faculty of Medicine and Health Sciences, Maulana Ibrahim Islamic State University of Malang, Malang 65144, Jawa Timur, Indonesia
| | - Zulvikar Syambani Ulhaq
- Research Center for Preclinical and Clinical Medicine, National Research and Innovation Agency Republic of Indonesia, Cibinong 16911, Indonesia
| |
Collapse
|
123
|
Chen G, Zhang Y, Zhou Y, Luo H, Guan H, An B. Targeting the mTOR Pathway in Hepatocellular Carcinoma: The Therapeutic Potential of Natural Products. J Inflamm Res 2024; 17:10421-10440. [PMID: 39659752 PMCID: PMC11630751 DOI: 10.2147/jir.s501270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/24/2024] [Indexed: 12/12/2024] Open
Abstract
Despite advancements in cancer treatment through surgery and drugs, hepatocellular carcinoma (HCC) remains a significant challenge, as reflected by its low survival rates. The mammalian target of rapamycin (mTOR) signaling pathway plays a crucial role in regulating the cell cycle, proliferation, apoptosis, and metabolism. Notably, dysregulation leading to the activation of the mTOR signaling pathway is common in HCC, making it a key focus for in-depth research and a target for current therapeutic strategies. This review focuses on the role of the mTOR signaling pathway and its downstream effectors in regulating HCC cell proliferation, apoptosis, autophagy, cell cycle, and metabolic reprogramming. Moreover, it emphasizes the potential of natural products as modulators of the mTOR signaling pathway. When incorporated into combination therapies, these natural products have been demonstrated to augment therapeutic efficacy and surmount drug resistance. These products target key signaling pathways such as mTOR signaling pathways. Examples include 11-epi-sinulariolide acetate, matrine, and asparagus polysaccharide. Their inhibitory effects on these processes suggest valuable directions for the development of more effective HCC therapeutic strategies. Various natural products have demonstrated the ability to inhibit mTOR signaling pathway and suppress HCC progression. These phytochemicals, functioning as mTOR signaling pathway inhibitors, hold great promise as potential anti-HCC agents, especially in the context of overcoming chemoresistance and enhancing the outcomes of combination therapies.
Collapse
Affiliation(s)
- Guo Chen
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Ya Zhang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Yaqiao Zhou
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Hao Luo
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Hongzhi Guan
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Baiping An
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
124
|
Wang M, Li T, Li W, Song T, Zhao C, Wu Q, Cui W, Hao Y, Hou Y, Zhu P. Unraveling the neuroprotective potential of scalp electroacupuncture in ischemic stroke: A key role for electrical stimulation. Neuroscience 2024; 562:160-181. [PMID: 39401739 DOI: 10.1016/j.neuroscience.2024.10.019] [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: 08/21/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024]
Abstract
This study aims to explore the neuroprotective effects of scalp Electroacupuncture (EA) on ischemic stroke, with a specific focus on the role of electrical stimulation (ES). Employing a rat model of middle cerebral artery occlusion (MCAO), we used methods such as Triphenyl tetrazolium chloride staining, micro-CT scanning, Enzyme linked immunosorbent assay (ELISA), and immunofluorescence to assess the impacts of EA. We further conducted RNA-seq analysis and in vitro experiments with organotypic brain slices and cerebral organoids to explore the underlying mechanisms. Our research revealed that EA notably reduced cerebral infarct volume and improved regional cerebral blood flow in rats following MCAO. Micro-CT imaging showed improved vascular integrity in EA-treated groups. Histological analyses, including HE staining, indicated reduced brain tissue damage. ELISA demonstrated a decrease in pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, suggesting improved blood-brain barrier function. Immunofluorescence and Western blot analyses revealed that EA treatment significantly inhibited microglial and astrocytic overactivation. RNA-seq analysis of brain tissues highlighted a downregulation of immune pathways and inflammatory responses, confirming the neuroprotective role of EA. This was further corroborated by in vitro experiments using organotypic brain slices and cerebral organoids, which showcased the efficacy of electrical stimulation in reducing neuroinflammation and protecting neuronal cells. The study highlights the potential of scalp EA, particularly its ES component, in treating ischemic stroke. It provides new insights into the mechanisms of EA, emphasizing its efficacy in neuroprotection and modulation of neuroinflammation, and suggests avenues for optimized treatment strategies in stroke therapy.
Collapse
Affiliation(s)
- Mingye Wang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Tongtong Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Wenyan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Tao Song
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Chi Zhao
- Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China
| | - Qiulan Wu
- Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China
| | - Wenwen Cui
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Yuanyuan Hao
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Yunlong Hou
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China; Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China; Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China; National Key Laboratory for Innovation and Transformation of Luobing Theory, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China; Key Laboratory of State Administration of TCM Cardio-Cerebral Vessel Collateral Disease, No.238, the South of Tianshan Street, Shijiazhuang, 050035, Hebei, China.
| | - Pengyu Zhu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, No. 411, The Street of Guogeli, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
125
|
Deng X, Zhang Q, Jin F, Lu F, Duan G, Han L, Zhu M, Yang Z, Zhang G. Ailanthone disturbs cross-talk between cancer cells and tumor-associated macrophages via HIF1-α/LINC01956/FUS/β-catenin signaling pathway in glioblastoma. Cancer Cell Int 2024; 24:397. [PMID: 39639311 PMCID: PMC11619249 DOI: 10.1186/s12935-024-03594-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND An increasing number of studies have focused on ailanthone (aila) due to its antitumor activity. However, the role of ailanthone in glioblastoma(GBM) has not been investigated before. This study aims to explore the biological function and the underlying mechanism of ailanthone in GBM. METHODS The microarray analysis was used to screen out down-stream long non-coding RNAs (lncRNAs) targeted by ailanthone. Real-time PCR(RT-PCR) assay was used to examine LINC01956 expression levels. Colony-formation, Methylthiazolyldiphenyl-tetrazolium bromide(MTT), cell-cycle, organoids culture and in-vivo tumorigenesis assays were used to examine cell growth in vitro and in vivo. Boyden assay was used to examine cell invasion ability in vitro. RNA immunoprecipitation and RNA-protein pull-down assays were used to examine the interaction between LINC01956 and FUS protein. Chromatin Immunoprecipitation(ChIP) assay was used to examine HIF1-α-binding sites in the LINC01956 promoter. RESULTS Ailanthone decreased GBM cell growth in vitro and in vivo via inducing ferroptosis. Ailanthone treatment exhibited blood‒brain barrier(BBB) permeability and specifically targeted the tumor area. LINC01956 was identified as a down-stream target of Ailanthone. LINC01956 exerted as an onco-lncRNA in GBM. M2 polarization of macrophages induced by exosomes derived from glioma cells overexpressing LINC01956 accelerated GBM progression. Mechanistically, we found that LINC01956 bound to FUS and reduced its ubiquitination. LINC01956 evoked nuclear translocation of phosphorylated (p)-β-catenin by recruiting FUS. Furthermore, under hypoxic conditions, LINC01956 was regulated by HIF-1α. Ailanthone decreased the expression of LINC01956 via suppressing HIF-1α. CONCLUSION Taken together, our data revealed for the first time that ailanthone regulated HIF-1α/LINC01956/FUS/β-catenin signaling pathway and thereby inhibited GBM progression.
Collapse
Affiliation(s)
- Xubin Deng
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qianbing Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Fa Jin
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangzhou, China
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fengfei Lu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangzhou, China
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guosheng Duan
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Luwei Han
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Meiling Zhu
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Zhengyan Yang
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China
| | - Gong Zhang
- The Radiotherapy Department of Shanxi Province People Hospital, Taiyuan, China.
| |
Collapse
|
126
|
Wang Y, Wang J, Liu J, Zhu H. Immune-related diagnostic markers for benign prostatic hyperplasia and their potential as drug targets. Front Immunol 2024; 15:1516362. [PMID: 39703506 PMCID: PMC11655502 DOI: 10.3389/fimmu.2024.1516362] [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: 10/24/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
Background Benign prostatic hyperplasia (BPH) is a common issue among older men. Diagnosis of BPH currently relies on imaging tests and assessment of urinary flow rate due to the absence of definitive diagnostic markers. Developing more accurate markers is crucial to improve BPH diagnosis. Method The BPH dataset utilized in this study was sourced from the Gene Expression Omnibus (GEO). Initially, differential expression and functional analyses were conducted, followed by the application of multiple machine learning techniques to identify key diagnostic markers. Subsequent investigations have focused on elucidating the functions and mechanisms associated with these markers. The ssGSEA method was employed to evaluate immune cell scores in BPH samples, facilitating the exploration of the relationship between key diagnostic markers and immune cells. Additionally, molecular docking was performed to assess the binding affinity of these key markers to therapeutic drugs for BPH. Tissue samples from BPH patients were collected for experimental validation of the expression differences of the aforementioned genes. Result A total of 185 differential genes were identified, comprising 67 up-regulated and 118 down-regulated genes. These genes are implicated in pathways that regulate extracellular matrix tissue composition and cellular responses to transforming growth factor beta stimulation, as well as critical signaling pathways such as AMPK and mTOR. Through the application of various machine learning techniques, DACH1, CACNA1D, STARD13, and RUNDC3B were identified as key diagnostic markers. The ssGSEA algorithm further corroborated the association of these diagnostic genes with diverse immune cells. Moreover, molecular docking analysis revealed strong binding affinities of these markers to tamsulosin and finasteride, suggesting their potential as drug targets. Finally, experimental validation confirmed the expression differences of DACH1, CACNA1D, STARD13, and RUNDC3B in BPH tissues. Conclusion This study introduces novel immune-related diagnostic markers for BPH and highlights their promise as new drug targets, providing a valuable approach for predictive diagnosis and targeted therapy of BPH.
Collapse
Affiliation(s)
- YaXuan Wang
- Cancer Research Centre Nantong, Affiliated Tumor Hospital of Nantong University & Nantong Tumor Hospital, Nantong, China
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Wang
- Department of Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jibin Liu
- Cancer Research Centre Nantong, Affiliated Tumor Hospital of Nantong University & Nantong Tumor Hospital, Nantong, China
| | - HaiXia Zhu
- Cancer Research Centre Nantong, Affiliated Tumor Hospital of Nantong University & Nantong Tumor Hospital, Nantong, China
| |
Collapse
|
127
|
Hao Y, Ji H, Gao L, Qu Z, Zhao Y, Chen J, Wang X, Ma X, Zhang G, Zhang T. Self-assembled carrier-free formulations based on medicinal and food active ingredients. Biomater Sci 2024; 12:6253-6273. [PMID: 39523875 DOI: 10.1039/d4bm00893f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The popularity of medicinal plants, which have a unique system and are mostly used in compound form for the prevention and treatment of a wide range of diseases, is growing worldwide. In recent years, with advances in chemical separation and structural analysis techniques, many of the major bioactive molecules of medicinal plants have been identified. However, the active ingredients in medicinal plants often possess chemical characteristics, including poor water solubility, stability and bioavailability, which limit their therapeutic applications. To address this problem, self-assembly of small molecules from medicinal food sources provides a new strategy. Driven by various types of acting forces, medicinal small molecules with modifiable groups, multiple sites of action, hydrophobic side chains, and rigid backbones with self-assembly properties are able to form various supramolecular network hydrogels, nanoparticles, micelles, and other self-assemblies. This review first summarizes the forms of self-assemblies such as supramolecular network hydrogels, nanoparticles, and micelles at the level of the action site, and discusses the recent studies on the active ingredients in medicinal plants that can be used for self-assembly, in addition to summarizing the advantages of self-assemblies for a variety of disease applications, including wound healing, antitumor, anticancer, and diabetes mellitus. Finally, the problems of self-assemblers and the possible directions for future development are presented. We firmly believe that self-assemblers have the potential to develop effective compounds from drug-food homologous plants, providing valuable information for drug research and new strategies and perspectives for the modernization of Chinese medicine.
Collapse
Affiliation(s)
- Yuan Hao
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Haixia Ji
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Li Gao
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Zhican Qu
- Shanxi Nanolattix Health Technology Co., Ltd, Taiyuan 030051, Shanxi, China
| | - Yinghu Zhao
- School of Environment and Safety Engineering, North University of China, Taiyuan 030051, Shanxi, China
| | - Jiahui Chen
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Xintao Wang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Xiaokai Ma
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Guangyu Zhang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Taotao Zhang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| |
Collapse
|
128
|
Li J, Ma X, Xu F, Yan Y, Chen W. Babaodan overcomes cisplatin resistance in cholangiocarcinoma via inhibiting YAP1. PHARMACEUTICAL BIOLOGY 2024; 62:314-325. [PMID: 38571483 PMCID: PMC10997361 DOI: 10.1080/13880209.2024.2331060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/05/2024]
Abstract
CONTEXT Cholangiocarcinoma with highly heterogeneous, aggressive, and multidrug resistance has a poor prognosis. Although babaodan (BBD) combined with cisplatin improved non-small cell lung cancer efficacy, its impact on overcoming resistance in cholangiocarcinoma remains unexplored. OBJECTIVE This study explored the role and mechanism of BBD on cisplatin resistance in cholangiocarcinoma cells (CCAs). MATERIALS AND METHODS Cisplatin-resistant CCAs were exposed to varying concentrations of cisplatin (25-400 μg/mL) or BBD (0.25-1.00 mg/mL) for 48 h. IC50 values, inhibition ratios, apoptosis levels, DNA damage, glutathione (GSH) levels, oxidized forms of GSH, total GSH content, and glutaminase relative activity were evaluated using the cell counting kit 8, flow cytometry, comet assay, and relevant assay kits. RESULTS BBD-reduced the cisplatin IC50 in CCAs from 118.8 to 61.83 μg/mL, leading to increased inhibition rate, apoptosis, and DNA damage, and decreased expression of B-cell lymphoma-2, p-Yes-associated protein 1/Yes-associated protein 1, solute carrier family 1 member 5, activating transcription factor 4, and ERCC excision repair 1 in a dose-dependent manner with maximum reductions of 78.97%, 51.98%, 54.03%, 56.59%, and 63.22%, respectively; bcl2-associated X and gamma histone levels were increased by 0.43-115.77% and 22.15-53.39%. The impact of YAP1 knockdown on cisplatin-resistant CCAs resembled BBD. GSH, oxidized GSH species, total GSH content, and glutaminase activity in cisplatin-resistant CCAs with BBD treatment also decreased, while YAP1 overexpression countered BBD's effects. DISCUSSION AND CONCLUSION This study provides a scientific basis for BBD clinical application and provides a new direction for BBD biological mechanism research.
Collapse
Affiliation(s)
- Jiong Li
- Department of Traditional Chinese Medicine, The First People’s Hospital of Lin’an District, Hangzhou, China
| | - Xiangjun Ma
- Department of Traditional Chinese Medicine, The First People’s Hospital of Lin’an District, Hangzhou, China
| | - Faying Xu
- College of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiqing Chen
- Department of General Surgery, The First People’s Hospital of Lin’an District, Hangzhou, China
| |
Collapse
|
129
|
Zeng M, Wang Y, Tao X, Fan T, Yin X, Shen C, Wang X. Novel Perspectives in the Management of Colorectal Cancer: Mechanistic Investigations Into the Reversal of Drug Resistance via Active Constituents Derived From Herbal Medicine. Phytother Res 2024; 38:5962-5984. [PMID: 39462152 DOI: 10.1002/ptr.8363] [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: 02/05/2024] [Revised: 09/03/2024] [Accepted: 10/05/2024] [Indexed: 10/29/2024]
Abstract
The high incidence and mortality rate of colorectal cancer have become a significant global health burden. Chemotherapy has been the traditional treatment for colorectal cancer and has demonstrated promising antitumor effects, leading to significant improvements in patient survival. However, the development of chemoresistance poses a major challenge during chemotherapy in colorectal cancer, significantly impeding treatment efficacy and affecting patient prognosis. Despite the development of a variety of novel anticolorectal cancer chemotherapy agents, their effectiveness and side effects vary, possibly due to the complex mechanisms of resistance in colorectal cancer. Abnormal drug metabolism or protein targets are the most direct causes of resistance. Further studies have revealed that these resistance mechanisms involve biochemical processes such as altered protein expression, autophagy, and epithelial-mesenchymal transitions. Herbal active ingredients offer an alternative treatment option and have shown promise in reversing colorectal cancer drug resistance. This paper aims to summarize the role of various biochemical processes and key protein targets in the occurrence and maintenance of resistance mechanisms in colorectal cancer. Additionally, it elaborates on the mechanisms of action of herbal active ingredients in reversing colorectal cancer drug resistance. The article also discusses the limitations and opportunities in developing novel anticolorectal cancer drugs based on herbal medicine.
Collapse
Affiliation(s)
- Mingtang Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelin Tao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Tianfei Fan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Yin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Shen
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Xueyan Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
130
|
Zheng ZX, Feng X, Zhuang L. The Effect of Oxidative Stress and Antioxidants Treatment on Gestational Diabetes Mellitus Outcome: A Scoping Review. Cell Biochem Biophys 2024; 82:3003-3013. [PMID: 39003362 DOI: 10.1007/s12013-024-01417-3] [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] [Accepted: 07/07/2024] [Indexed: 07/15/2024]
Abstract
Diagnosing a pregnant woman's glucose intolerance is referred to as gestational diabetes mellitus (GDM). Diabetes has been linked to enhanced oxidative stress. In this condition, oxidative stress may damage nucleic acids, fats, and proteins, which in turn affects cell and tissue functions. The present study highlights the relationship between oxidative stress and GDM, with a particular focus on the role of hyperglycemia-induced processes during reactive oxygen species (ROS) oversupply, followed by it discusses the oxidative stress biomarkers and assesses the effects of antioxidant supplements on glycemic control, inflammatory processes, and oxidative stress among individuals with GDM. Two reviewers conducted a comprehensive literature search utilizing the PubMed®, Web of Science™, and Scopus® databases. Only items published in the English language up until June 2024 were taken into account. We conducted a thorough search of research databases to identify articles that had the terms "oxidative stress" or "antioxidant" and "GDM". From this search, we selected 55 relevant papers to be included in this narrative review. Pregnancy-induced hypertension, postpartum bleeding, lower birth weight, a higher risk of hyperbilirubinemia in their neonates, fetal growth retardation, and birth asphyxia were revealed to be outcomes of women enduring major oxidative stress during pregnancy. Furthermore, tight glycemic control both before and throughout pregnancy as well as oxidative stress treatment may help women highly prone to GDM.
Collapse
Affiliation(s)
- Zhen-Xia Zheng
- Obstetrics Department, Zhongshan Hospital of Xiamen University, Fujian, China
| | - Xiao Feng
- Department of Pediatrics, Affiliated Zhongshan Hospital of Xiamen University, Fujian, China
| | - Lijuan Zhuang
- Department of Obstetrics and Gynecology, Zhongshan Hospital of Xiamen University, Fujian, China.
| |
Collapse
|
131
|
Wu Y, Lin Y, Liu B, Ma J, Xiang Y, Wang Y, Meng S. Shexiang Tongxin dropping pill ameliorates microvascular obstruction via downregulating ALOX12 after myocardial ischemia-reperfusion. Int J Cardiol 2024; 416:132481. [PMID: 39179033 DOI: 10.1016/j.ijcard.2024.132481] [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: 03/29/2024] [Revised: 08/04/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Microvascular dysfunction (MVD) is common in patients with myocardial infarction receiving reperfusion therapy and is associated with adverse cardiac prognosis. Accumulating evidence suggests a protective role of Shexiang Tongxin dropping pill (STDP) in MVD. However, the specific effects and the underlying mechanisms of STDP in the context of MVD after myocardial ischemia-reperfusion (IR) remains unclear. AIMS We aimed to elucidate the role of STDP in MVD induced by IR and the potential mechanisms involved. METHODS Mice were orally administered with STDP or normal saline for 5 days before receiving myocardial IR. Cardiac function and microvascular obstruction was measured. Proteomics and single-cell RNA sequencing was performed on mouse hearts. In vitro hyoxia/reoxygenation model was established on mouse cardiac microvascular endothelial cells (MCMECs). RESULTS STDP improved cardiac function and decreased microvascular obstruction (MVO) in mice after myocardial IR. Proteomics identified ALOX12 as an important target of STDP. Single-cell RNA sequencing further revealed that downregulation of ALOX12 by STDP mainly occurred in endothelial cells. The involvement of ALOX12 in the effect of STDP on MVO was validated by manipulating ALOX12 via endothelial-specific adeno-associated virus transfection in vivo and in vitro. In vivo, overexpression of ALOX12 increased whereas knockdown of ALOX12 decreased MVO and thrombus formation. STDP treatment alleviated the detrimental effects of overexpression of ALOX12. In vitro, overexpression of ALOX12 increased endothelial cell inflammation and platelet adhesion to endothelial cells, which was abolished by STDP treatment. CONCLUSION Our findings suggest that STDP alleviates MVO after IR, with ALOX12 playing a crucial role.
Collapse
Affiliation(s)
- Yuanhao Wu
- Medical School Of Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China
| | - Yanjun Lin
- Medical School Of Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China; Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China
| | - Bo Liu
- Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China
| | - Jingqing Ma
- Medical School Of Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China
| | - Yin Xiang
- Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China
| | - Yuepeng Wang
- Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China.
| | - Shu Meng
- Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China.
| |
Collapse
|
132
|
Zeng Y, Wang Z, Zhang J, Jian W, Fu Q. Antitumour activity of oleanolic acid: A systematic review and meta‑analysis. Oncol Lett 2024; 28:582. [PMID: 39421313 PMCID: PMC11484195 DOI: 10.3892/ol.2024.14715] [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: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Oleanolic acid (OA), a compound known for its potent antitumour properties, has been the subject of investigations in both cell and animal models. Although OA has good biological activity, its low water solubility and bioavailability limit its therapeutic use, and therefore translating the potential of OA into the clinical oncology setting remains challenging. The present systematic review and meta-analysis utilized evidence from animal model studies to gain insights into the antitumour mechanisms of OA to address the gap in understanding, and to provide guidance for future research directions and potential clinical applications. The guidelines outlined in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses were applied in the present study and a comprehensive search was conducted across the PubMed/MEDLINE, Web of Science, Cochrane Library and Embase databases, with a cut-off date of June 30, 2023. The primary focus was on randomized controlled trials that used animal models to assess the antitumour effects of OA. The methodological quality appraisal was conducted using the Systematic Review Centre for Laboratory Animal Experimentation risk of bias tool, and tumour volume and weight served as the principal outcome measures. Data were analysed using the RevMan (version 5.3) and Stata SE11 software packages, with an assessment of heterogeneity conducted using the I2 statistical test, sensitivity analysis conducted using the leave-one-out approach, and evaluation of publication bias performed using Egger's test and funnel plot analysis. The present study demonstrated a significant inhibitory effect of OA intervention on tumour growth and a decrease in tumour weight in animal models. Despite the broad spectrum of antitumour effects exhibited by OA, further investigations are warranted to optimize the dosage and administration routes of OA to maximize its efficacy in clinical cancer treatment.
Collapse
Affiliation(s)
- Ying Zeng
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Zhonglian Wang
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Jing Zhang
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Wei Jian
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Qiaofen Fu
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
133
|
Chen YY, Zeng XT, Gong ZC, Zhang MM, Wang KQ, Tang YP, Huang ZH. Euphorbia Pekinensis Rupr. sensitizes colorectal cancer to PD-1 blockade by remodeling the tumor microenvironment and enhancing peripheral immunity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156107. [PMID: 39368338 DOI: 10.1016/j.phymed.2024.156107] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/28/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Immune checkpoint blockade, such as monoclonal antibodies targeting programmed cell death protein 1 (PD-1), has been a major breakthrough in the treatment of several cancers, but has limited effect in colorectal cancer (CRC), which is a highly prevalent cancer worldwide. Current chemotherapy-based strategies to boost PD-1 response have many limitations. And the role of peripheral immunity in boosting PD-1 response continues to attract attention. Therefore, candidate combinations of PD-1 blockade need to be drugs with multi-targets and multi-modulatory functions. However, it is still unknown whether traditional Chinese medicines with such property can enhance the applicability and efficacy of PD-1 blockade in colorectal cancer. METHODS Euphorbia Pekinensis extract (EP) was prepared and the constituents were analyzed by HPLC. CRC cells were used for in vitro experiments, including cell viability assay, colony formation assay, flow cytometry for 7-AAD staining, western blotting for caspase 3 and caspase 7, HMGB1 and ATP detection. An orthotopic CT26 mouse model was subsequently used to investigate the combination of EP and PD-1 blockade therapy. Tumor volume and tumor weight were assessed, tumor tissues were subjected to histopathological HE staining and TUNEL staining, and tumor-infiltrating immune cells were evaluated by immunofluorescence staining. RNA-sequencing, target prediction and pathway analysis were further employed to explore the mechanism. Molecular docking and cellular thermal shift assay (CETSA) were utilized to verify the direct target of the core component of EP. And, loss-of-function analysis was carried to confirm the upstream-downstream relationship. Flow cytometry was employed to analyze CD8+ T cells in the peripheral blood and spleen. RESULTS The main constituents of EP are diterpenoids and flavonoids. EP dramatically suppresses CRC cell growth and exerts its cytotoxic effect by triggering immunogenic cell death in vitro. Moreover, EP synergizes with PD-1 blockade to inhibit tumorigenesis in tumor-bearing mice. Disruption of ISX nuclear localization by helioscopinolide E is a central mechanism of EP-induced apoptosis in CRC cell. Meanwhile, EP activates immune response by upregulating Phox2b to reshape the immune microenvironment. In addition, EP regulates peripheral immunity by regulating the T cell activation and proliferation, and the ratio of CD8+ T cells in peripheral blood is drastically increased, thereby enhancing the therapeutic efficacy of anti-PD1 immunotherapy. CONCLUSION EP triggers intra-tumor immunogenic cell death and modulates the immunoregulatory signaling to elicit the tumor immunogenicity. Moreover, EP participates in transcriptional activation of immune response-related pathways. Consequently, multiple stimulating functions of EP on macro- and micro-immune potentiates the anti-tumor effect of PD-1 blockade in CRC.
Collapse
Affiliation(s)
- Yan-Yan Chen
- Wuxi Cancer Institute, Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiao-Tao Zeng
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, China
| | - Zhi-Cheng Gong
- Wuxi Cancer Institute, Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Mei-Mei Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, China
| | - Kai-Qing Wang
- Wuxi Cancer Institute, Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, China.
| | - Zhao-Hui Huang
- Wuxi Cancer Institute, Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
134
|
Han MM, Fan YK, Zhang Y, Dong ZQ. Advances in herbal polysaccharides-based nano-drug delivery systems for cancer immunotherapy. J Drug Target 2024; 32:311-324. [PMID: 38269853 DOI: 10.1080/1061186x.2024.2309661] [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: 10/13/2023] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
The boom in cancer immunotherapy has provided many patients with a better chance of survival, but opportunities often come with challenges. Single immunotherapy is not good enough to eradicate tumours, and often fails to achieve the desired therapeutic effect because of the low targeting of immunotherapy drugs, and causes more side effects. As a solution to this problem, researchers have developed several nano Drug Delivery Systems (NDDS) to deliver immunotherapeutic agents to achieve good therapeutic outcomes. However, traditional drug delivery systems (DDS) have disadvantages such as poor bioavailability, high cytotoxicity, and difficulty in synthesis, etc. Herbal Polysaccharides (HPS), derived from natural Chinese herbs, inherently possess low toxicity. Furthermore, the biocompatibility, biodegradability, hydrophilicity, ease of modification, and immunomodulatory activities of HPS offer unique advantages in substituting traditional DDS. This review initially addresses the current developments and challenges in immunotherapy. Subsequently, it focuses on the immunomodulatory mechanisms of HPS and their design as nanomedicines for targeted drug delivery in tumour immunotherapy. Our findings reveal that HPS-based nanomedicines exhibit significant potential in enhancing the efficacy of cancer immunotherapy, providing crucial theoretical foundations and practical guidelines for future clinical applications.
Collapse
Affiliation(s)
- Miao-Miao Han
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yi-Kai Fan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- Joint Research Center for Chinese Medicinal Herbs, IMPLAD, ABRC & ACCL, Beijing, China
| | - Zheng-Qi Dong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- Joint Research Center for Chinese Medicinal Herbs, IMPLAD, ABRC & ACCL, Beijing, China
| |
Collapse
|
135
|
Tan XD, Luo CF, Liang SY. Antihyperlipidemic drug rosuvastatin suppressed tumor progression and potentiated chemosensitivity by downregulating CCNA2 in lung adenocarcinoma. J Chemother 2024; 36:662-674. [PMID: 38288951 DOI: 10.1080/1120009x.2024.2308975] [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: 10/17/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 11/22/2024]
Abstract
Rosuvastatin (RSV) is widely used to treat hyperlipidemia and hypercholesterolemia and is recommended for the primary and secondary prevention of cardiovascular diseases (CVD). In this study, we aimed to explore its action and mechanism in lung adenocarcinoma (LUAD) therapy. Lewis and CMT64 cell-based murine subcutaneous LUAD models were employed to explore the effects of RSV monotherapy combined with cisplatin and gemcitabine. Human lung fibroblasts and human LUAD cell lines were used to assess the effects of RSV on normal and LUAD cells. Bioinformatics and RNA interference were used to observe the contribution of cyclin A2 (CCNA2) knockdown to RSV inhibition and to improve chemosensitivity in LUAD. RSV significantly suppressed grafted tumor growth in a murine subcutaneous LUAD model and exhibited synergistic anti-tumor activity with cisplatin and gemcitabine. In vitro and in vivo experiments demonstrated that RSV impaired the proliferation and migration of cancer cells while showing little inhibition of normal lung cells. RNA interference and CCK8 detection preliminarily indicated that RSV inhibited tumor growth and enhanced the chemosensitivity to cisplatin and gemcitabine by downregulating CCNA2. RSV suppressed LUAD progression and enhanced chemosensitivity to cisplatin and gemcitabine by downregulating CCNA2, which should be prior consideration for the treatment of LUAD, especially for patients co-diagnosed with hyperlipidemia and hypercholesterolemia.
Collapse
Affiliation(s)
- Xiang-Di Tan
- The Fourth Affiliated Hospital, Guangzhou Medical University, Zengcheng, China
| | - Cui-Fang Luo
- The Fourth Affiliated Hospital, Guangzhou Medical University, Zengcheng, China
| | - Si-Yu Liang
- The Fourth Affiliated Hospital, Guangzhou Medical University, Zengcheng, China
| |
Collapse
|
136
|
Huang C, Lan H, Bai M, Chen J, Xu S, Sun Q, Chen Q, Mao W, Jiang J, Zhu J. Rifaximin alleviates irinotecan-induced diarrhea in mice model. Ann Med 2024; 56:2429029. [PMID: 39575573 PMCID: PMC11587719 DOI: 10.1080/07853890.2024.2429029] [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: 01/21/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Irinotecan is a chemotherapeutic drug widely used to treat solid tumors. However, its effectiveness is limited by the severely delayed onset of diarrhea. This study aimed to confirm the protective effects of the non-systemic oral antibiotic rifaximin on irinotecan-induced mucositis in mice model. MATERIALS AND METHODS Six to eight week-old BALB/c mice were treated with saline, irinotecan (50 mg/kg, i.p. once daily), rifaximin (50 mg/kg, p.o. twice daily), or irinotecan + rifaximin for 9 consecutive days. Signs of diarrhea, bloody diarrhea, and body weight were monitored daily. Intestinal tissues were harvested for histopathological analysis and quantitative PCR. SN38 and SN38G concentration in intestine were detected using LC-MS analysis. Intestinal bacteria β-glucuronidase (BGUS) activity was detected using mouse feces. We performed 16S rRNA sequencing to investigate the gut microbiota composition. Gut permeability was tested in vivo by measuring the fluorescein isothiocyanate-dextran intensity in the serum. RESULTS Rifaximin reduced the frequency of delayed diarrhea and attenuated the severity of diarrhea caused by irinotecan in mice. Rifaximin significantly inhibited SN38 exposure in intestine and irinotecan-induced increase in BGUS activity. Rifaximin alleviated intestinal mucosal inflammation, prevented intestinal epithelial damage caused by irinotecan, and maintained gut barrier function. Moreover, the consecutive use of rifaximin did not cause a disorder in gut microbiota and reduced irinotecan-induced Firmicutes expansion. More importantly, rifaximin inhibited the expansion of some microbiota (such as Blautia, Eggerthella, and f_Enterobacteriaceae) and promoted an increase in beneficial microbiota (such as Lactobacillus intestinalis, Lachnospiraceae NK4A136 group, and f_Oscillospiraceae). CONCLUSIONS Preventive use of rifaximin is a feasible method to protect against irinotecan-induced diarrhea.
Collapse
Affiliation(s)
- Chengyi Huang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Huiyin Lan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Minghua Bai
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jinggang Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Shengkun Xu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Quanquan Sun
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jin Jiang
- Department of Oncology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
137
|
Kim J, An J, Song Y, Jang M, Kong H, Kim S. Effect of Elderberry ( Sambucus nigra L.) Extract Intake on Normalizing Testosterone Concentration in Testosterone Deficiency Syndrome Rat Model Through Regulation of 17β-HSD, 5α-Reductase, and CYP19A1 Expression. Nutrients 2024; 16:4169. [PMID: 39683564 DOI: 10.3390/nu16234169] [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: 10/20/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives. Men experience Leydig cell and mitochondrial dysfunction due to the accumulation of reactive oxygen species during aging, leading to hormonal imbalances in the body. This results in symptoms of testosterone deficiency syndrome (TDS) as testosterone levels decline. Consequently, there is a growing need for alternative therapies, such as phytotherapy, to regulate testosterone secretion. Methods. In this study, we evaluated the potential of elderberry extract powder (KSB191) as a functional ingredient for improving TDS by analyzing its mechanism in regulating testosterone imbalance. The major compounds of KSB191 were rutin and fructose-leucine, and the efficacy of KSB191 was confirmed by observing increases in total testosterone, free testosterone, and sperm motility in an aged rat model with decreased testosterone levels. Additionally, we assessed safety by analyzing levels of prostate-specific antigen, alanine aminotransferase, aspartate aminotransferase, and creatinine. Results. To confirm the effectiveness of KSB191 in increasing testosterone synthesis and inhibiting its breakdown, we analyzed the expression levels of genes related to testosterone synthesis and degradation in the testis tissue. KSB191 not only increases the expression levels of enzymes (3β-HSD, CYP17A1, and 17β-HSD) that catalyze testosterone synthesis in Leydig cells, but also reduces the expression of enzymes (5α-reductase and CYP19A1) that degrade testosterone, thereby enhancing testosterone production in the body. Conclusions. KSB191 is predicted to be a novel functional ingredient that acts on Leydig cells and increases testosterone synthesis (particularly, the increase in free testosterone), ultimately alleviating the symptoms of TDS.
Collapse
Affiliation(s)
- Jiyeon Kim
- KOSABIO Inc., Namyangju-si 12106, Republic of Korea
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Jinho An
- KOSABIO Inc., Namyangju-si 12106, Republic of Korea
| | - Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Mincheol Jang
- Hurum Co., Ltd., Geumcheon-gu, Seoul 08505, Republic of Korea
| | - Hyunseok Kong
- Department of Animal Science, Sahmyook University, Seoul 01795, Republic of Korea
- PADAM Natural Material Research Institute, Sahmyook University, Seoul 01795, Republic of Korea
| | - Sangbum Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
138
|
Li S, Dai A, Zhou Y, Chen X, Chen Y, Zhou L, Yang X, Yue M, Shi J, Qiu Y. Efficacy of combination scalp acupuncture for post-stroke cognitive impairment: a systematic review and meta-analysis. Front Neurosci 2024; 18:1468331. [PMID: 39678533 PMCID: PMC11638195 DOI: 10.3389/fnins.2024.1468331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Objective This systematic review and meta-analysis aimed to evaluate the efficacy of combination scalp acupuncture in treating post-stroke cognitive impairment. Methods A comprehensive search was conducted across eight databases: PubMed, Web of Science, Cochrane Database, Embase, CBM, CNKI, WanFang, and VIP, targeting randomized controlled trials (RCTs) published from the inception of these databases until October 24, 2024. The inclusion criteria focused on RCTs that compared scalp acupuncture with conventional treatments as therapeutic interventions for patients suffering from post-stroke cognitive impairment (PSCI). The effectiveness of these treatments was evaluated using various outcome measures, including the Mini-Mental State Examination (MMSE), the Montreal Cognitive Assessment (MoCA), the Loewenstein Occupational Therapy Cognitive Assessment (LOTCA), as well as P300 latency and amplitude, which collectively assess cognitive function. Two independent reviewers conducted a risk of bias (ROB2) assessment, and data analysis was performed using Review Manager (RevMan) version 5.4. Results This analysis included a total of 28 studies involving 1,995 patients. However, according to the standards of the ROB2 tool, most of these studies exhibited various methodological issues. The comprehensive analysis indicates that the efficacy of combined scalp acupuncture in treating post-stroke cognitive impairment (PSCI) is superior to that of single treatments, as evidenced by improvements across multiple scales, including the Montreal Cognitive Assessment (MoCA), Mini-Mental State Examination (MMSE), Loewenstein Occupational Therapy Cognitive Assessment (LOTCA), P300 latency, and amplitude. Specifically, the overall effective rate was reported as (RR = 1.28, 95% CI: 1.14-1.45, p < 0.0001; I 2 = 51%, random effects model). The mean differences for the various scales were as follows: MoCA (MD = 3.55, 95% CI: 2.68-4.41, I 2 = 93%, random effects model), MMSE (MD = 3.78, 95% CI: 2.83-4.73, I 2 = 94%, random effects model), LOTCA (MD = 9.70, 95% CI: 7.72-11.69, I 2 = 57%, random effects model), P300 latency (MD = -21.83, 95% CI: -26.31 to -17.35, I 2 = 55%, random effects model), and amplitude (MD = 1.05, 95% CI: 0.76-1.34, I 2 = 0%, fixed effects model), demonstrating low, medium, and high levels of heterogeneity, respectively. Notably, one study reported an adverse event related to participant withdrawal during the study. Conclusion Combination scalp acupuncture exhibits superior efficacy compared to single-treatment modalities in patients with post-stroke cognitive impairment (PSCI). However, the higher risk of bias (ROB) in the included trials suggests that the quality of evidence about these assessment results may be compromised. Therefore, there is an urgent need for additional high-quality clinical trials to further validate the efficacy and effectiveness of combined scalp acupuncture in treating PSCI, ultimately enhancing the overall level of evidence. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024519200.
Collapse
Affiliation(s)
- Song Li
- Yunnan University of Traditional Chinese Medicine, Kunming, China
- Department of Acupuncture and Moxibustion, Yunnan Provincial Hospital of Traditional, Chinese Medicine, The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, China
| | - Anhong Dai
- Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Yihao Zhou
- Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xu Chen
- Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yizhou Chen
- Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Li Zhou
- Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Xiaolin Yang
- Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Mengqi Yue
- Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Jing Shi
- Yunnan University of Traditional Chinese Medicine, Kunming, China
- Department of Acupuncture and Moxibustion, Yunnan Provincial Hospital of Traditional, Chinese Medicine, The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, China
| | - Yong Qiu
- Yunnan University of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
139
|
Li Y, Chen H, Zhao Y, Yan Q, Chen L, Song Q. circUBE2G1 interacts with hnRNPU to promote VEGF-C-mediated lymph node metastasis of lung adenocarcinoma. Front Oncol 2024; 14:1455909. [PMID: 39664183 PMCID: PMC11631705 DOI: 10.3389/fonc.2024.1455909] [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: 06/27/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
Background Patients with lymph node(LN)metastasis-positive Lung adenocarcinoma(LUAD)suffer from a significantly reduced five-year survival rate. Increasing evidence indicates circular RNAs(circRNAs)play crucial roles in regulating cancer progression. However, the specific regulatory mechanisms of circRNAs in the LN metastasis of LUAD have not been fully explored. Methods GEO datasets and sequence analysis were applied for the identification of differentially expressed circRNAs between LUAD tissues and adjacent normal tissues. In vitro and in vivo experiments were performed to evaluate the function of circUBE2G1. The interaction between circUBE2G1 and VEGF-C was determined by RNA pulldown, ChIP, ChIRP and luciferase assays. Results In this study, we identified a novel circRNA, circUBE2G1 (hsa_circ_0041555), which is upregulated in LUAD and positively correlated with LN metastasis in patients with LUAD. Functionally, overexpression of circUBE2G1 promotes lymphangiogenesis and LN metastasis of LUAD both in vitro and in vivo. Mechanistically, circUBE2G1 activates the transcription of vascular endothelial growth factor C (VEGF-C) by recruiting hnRNPU to enhance H3K27ac on the VEGF-C promoter, thereby facilitating lymphangiogenesis and LN metastasis in LUAD. Conclusion Our findings offer new insights into the mechanisms behind circRNA-mediated LN metastasis in LUAD and suggest that circUBE2G1 may serve as a potential therapeutic target for LN metastasis in LUAD.
Collapse
Affiliation(s)
- Yuting Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hui Chen
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Yue Zhao
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qilu Yan
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
140
|
Zhou M, Chen Y, Jin W, Li P, Hu J, Guo X. Traditional Chinese Medicine: A Promising Treatment Option for Intestinal Fibrosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2107-2129. [PMID: 39581857 DOI: 10.1142/s0192415x24500812] [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: 11/26/2024]
Abstract
Intestinal fibrosis, a common complication of inflammatory bowel disease, in particular in Crohn's disease, arises from chronic inflammation, leading to intestinal narrowing, structural damage, and functional impairment that significantly impact patients' quality of life. Current treatment options for intestinal fibrosis are limited, with surgery being the primary intervention. Traditional Chinese Medicine (TCM) has emerged as a promising approach in preventing and treating intestinal fibrosis. However, there is a scarcity of literature summarizing the mechanisms underlying TCM's efficacy in this context. To address this gap, we conducted a comprehensive review, uncovering multiple mechanisms through which TCM mitigates intestinal fibrosis. These mechanisms include immune cell balance regulation, suppression of inflammatory responses, reduction of inflammatory mediators, alleviation of colon tissue damage, restoration of intestinal function, modulation of growth factors to inhibit fibroblast activation, dynamic regulation of TIMPs and MMPs to reduce extracellular matrix deposition, inhibition of epithelial-mesenchymal transition and endothelial-mesenchymal transition, autophagy modulation, maintenance of the intestinal mucosal barrier, prevention of tissue damage by harmful factors, and regulation of cell proliferation and apoptosis. This study aims to bridge existing knowledge gaps by presenting recent evidence supporting the utilization of TCM in both clinical and experimental research settings.
Collapse
Affiliation(s)
- Meng'en Zhou
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Yan Chen
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Wenqi Jin
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Peng Li
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Jie Hu
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Xiutian Guo
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| |
Collapse
|
141
|
Sun X, Shukla M, Wang W, Li S. Unlocking gut-liver-brain axis communication metabolites: energy metabolism, immunity and barriers. NPJ Biofilms Microbiomes 2024; 10:136. [PMID: 39587086 PMCID: PMC11589602 DOI: 10.1038/s41522-024-00610-9] [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: 08/07/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
The interaction between the gut-microbiota-derived metabolites and brain has long been recognized in both health and disease. The liver, as the primary metabolic organ for nutrients in animals or humans, plays an indispensable role in signal transduction. Therefore, in recent years, Researcher have proposed the Gut-Liver-Brain Axis (GLBA) as a supplement to the Gut-Brain Axis. The GLBA plays a crucial role in numerous physiological and pathological mechanisms through a complex interplay of signaling pathways. However, gaps remain in our knowledge regarding the developmental and functional influences of the GLBA communication pathway. The gut microbial metabolites serve as communication agents between these three distant organs, functioning prominently within the GLBA. In this review, we provide a comprehensive overview of the current understanding of the GLBA, focusing on signaling molecules role in animal and human health and disease. In this review paper elucidate its mechanisms of communication, explore its implications for immune, and energy metabolism in animal and human, and highlight future research directions. Understanding the intricate communication pathways of the GLBA holds promise for creating innovative treatment approaches for a wide range of immune and metabolic conditions.
Collapse
Affiliation(s)
- Xiaoge Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Manish Shukla
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.
| |
Collapse
|
142
|
Yuan W, Zhang J, Chen H, Zhuang Y, Zhou H, Li W, Qiu W, Zhou H. Natural compounds modulate the mechanism of action of tumour-associated macrophages against colorectal cancer: a review. J Cancer Res Clin Oncol 2024; 150:502. [PMID: 39546016 PMCID: PMC11568041 DOI: 10.1007/s00432-024-06022-8] [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: 09/11/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Colorectal cancer (CRC) exhibits a substantial morbidity and mortality rate, with its aetiology and pathogenesis remain elusive. It holds significant importance within the tumour microenvironment (TME) and exerts a crucial regulatory influence on tumorigenesis, progression, and metastasis. TAMs possess the capability to foster CRC pathogenesis, proliferation, invasion, and metastasis, as well as angiogenesis, immune evasion, and tumour resistance. Furthermore, TAMs can mediate the prognosis of CRC. In this paper, we review the mechanisms by which natural compounds target TAMs to exert anti-CRC effects from the perspective of the promotional effects of TAMs on CRC, mainly regulating the polarization of TAMs, reducing the infiltration and recruitment of TAMs, enhancing the phagocytosis of macrophages, and regulating the signalling pathways and cytokines, and discuss the potential value and therapeutic strategies of natural compounds-targeting the TAMs pathway in CRC clinical treatment.
Collapse
Affiliation(s)
- Weichen Yuan
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiexiang Zhang
- Urology Centre, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Surgery of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haibin Chen
- Science and Technology Department, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yupei Zhuang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongli Zhou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenting Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Wenli Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Hongguang Zhou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
143
|
Yan J, Liu H, Shang J, Fang Q, Ye J, Lu X, Fan X. Protective effects of Shexiang-Tongxin dropping pill against acute myocardial infarction in rats through inhibition of apoptosis and ERK/MAPK signaling pathways. Heliyon 2024; 10:e39939. [PMID: 39553562 PMCID: PMC11565526 DOI: 10.1016/j.heliyon.2024.e39939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
Acute myocardial infarction (AMI) remains a significant health challenge globally, highlighting the ongoing need for effective treatments. Shexiang-Tongxin dropping pill (STDP) is widely utilized as a therapeutic option for AMI in China and Southeast Asia. However, the intricate mechanisms of action of STDP against AMI remain largely unknown. The pharmacodynamic effects of STDP in treating AMI were evaluated both in vitro and in vivo using human umbilical vein endothelial cell oxygen-glucose deprivation, RAW264.7 cell inflammatory injury, and rat left anterior descending surgery models. The whole transcriptome sequencing was performed to analyze gene expression changes in experimental rat hearts after left anterior descending surgery. An integrative approach combining network pharmacology and sequencing data was used to determine the multi-target and multi-pathway mechanisms underlying the action of STDP against AMI. Molecular docking was conducted to identify the primary anti-AMI ingredients in STDP. STDP treatment significantly resisted AMI in vivo and protected against inflammatory and hypoxic injuries in vitro. It resulted in 63 % (901 of 1430) of genes showing restorative regulation in the AMI disease network, relating to the TGF-β, PI3K, apoptosis, and MAPK pathways. Validation experiments indicated that inhibiting apoptosis and ERK/MAPK pathways by reducing Bax and p-ERK1/2 expression levels in rat hearts may be a crucial mechanism of STDP against AMI. Molecular target prediction indicated that tanshinone IIA, salvianolic acid A, salvianolic acid B, and resibufogenin were the essential pharmacodynamic substances of STDP in AMI treatment. This study sheds light on novel mechanisms by which STDP rebalances the AMI disease network through its multi-target and multi-pathway effects. The findings offer data support for the more precise clinical application of STDP.
Collapse
Affiliation(s)
- Jun Yan
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hanbing Liu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiaxin Shang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qianqian Fang
- Inner Mongolia Conba Pharmaceutical Co., Ltd., Ordos, 017000, China
| | - Jianfeng Ye
- Inner Mongolia Conba Pharmaceutical Co., Ltd., Ordos, 017000, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, 314100, Jiaxing, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Xiaohui Fan
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, 314100, Jiaxing, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| |
Collapse
|
144
|
Liu C, Lin X, Huang M, Zhang S, Che L, Lai Z, Chen X, Pu W, Yang S, Qiu Y, Yu H. Babaodan inhibits cell proliferation and metastasis and enhances anti-tumor effects of camrelizumab by inhibiting M2 phenotype macrophages in hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118540. [PMID: 38992397 DOI: 10.1016/j.jep.2024.118540] [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: 02/01/2024] [Revised: 05/20/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Babaodan (BBD) is a unique Chinese medication utilized in traditional Chinese medicine. It can eliminate toxins, induce diuresis, and eliminate yellowish hue. In addition to treating acute and chronic viral hepatitis, cholecystitis, cholangitis, and urinary tract infections, BBD has garnered popularity as a substitution treatment for several malignant cancers, particularly hepatocellular carcinoma (HCC). AIM OF THE STUDY To elucidate the efficacy and mechanism of BBD alone and combined with camrelizumab (CLM) for treating HCC. METHODS We investigated the effects of BBD on the HCC tumor microenvironment in vivo. Furthermore, we evaluated its effects on tumor growth and metastasis induced by M2 macrophages in vitro. RESULTS In a mouse model of orthotopic HCC, BBD decreased tumor growth. Furthermore, it increased the M1/M2 macrophage ratio and CD8+ T-cell abundance in mice. In addition, BBD reversed HCC cell proliferation and metastasis induced by M2 macrophages, increased the anti-HCC effect of low-dose CLM, and attenuated organ damage induced by high-dose CLM. Lastly, BBD enhanced the efficacy of CLM via the PI3K/AKT/mTOR signaling pathway. CONCLUSION BBD increases the antitumor effect of CLM by modulating the tumor immune microenvironment and attenuating its the toxic side effects of CLM.
Collapse
Affiliation(s)
- Caiyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaowei Lin
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Manru Huang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Siqi Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Li Che
- Xiamen Traditional Chinese Medicine Co., Ltd., Xiamen, 361100, China
| | - Zhicheng Lai
- Xiamen Traditional Chinese Medicine Co., Ltd., Xiamen, 361100, China
| | - Xiyi Chen
- Xiamen Traditional Chinese Medicine Co., Ltd., Xiamen, 361100, China
| | - Weiling Pu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shenshen Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
145
|
Sharma R, Mehan S, Khan Z, Das Gupta G, Narula AS. Therapeutic potential of oleanolic acid in modulation of PI3K/Akt/mTOR/STAT-3/GSK-3β signaling pathways and neuroprotection against methylmercury-induced neurodegeneration. Neurochem Int 2024; 180:105876. [PMID: 39368746 DOI: 10.1016/j.neuint.2024.105876] [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: 06/25/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that gradually deteriorates motor neurons, leading to demyelination, muscle weakness, and eventually respiratory failure. The disease involves several pathological processes, such as increased glutamate levels, mitochondrial dysfunction, and persistent neuroinflammation, often exacerbated by environmental toxins like mercury. This study explores the therapeutic potential of Olea europaea active phytoconstituents oleanolic acid (OLA) against ALS by targeting the overactivated PI3K/Akt/mTOR/STAT-3/GSK-3β signalling pathways. Methods involved in-silico studies, in vitro and in vivo experiments in which varying doses of methylmercury 5 mg/kg, p.o. and OLA (100 and 200 mg/kg, i.p.) were administered to rats for 42 days. Behavioural assessments, gross morphological, histopathological, and neurochemical parameters were measured in cerebrospinal fluid (CSF), blood plasma, and brain homogenates (cerebral cortex, hippocampus, striatum, midbrain, cerebellum) along with complete blood count (CBC) analysis. Results revealed OLA's significant neuroprotective properties. OLA effectively modulated targeted pathways, reducing pro-inflammatory cytokines, restoring normal levels of myelin basic protein (MBP) and neurofilament light chain (NEFL), and reducing histopathological changes. Gross pathological studies indicated less tissue damage, while CBC analysis showed improved hematology parameters. Additionally, the combination of OLA and edaravone (10 mg/kg, i.p.) demonstrated enhanced efficacy, improving motor functions and extending survival in ALS model rats. In conclusion, OLA exhibits significant therapeutic potential for ALS, acting as a potent modulator of key pathological signaling pathways. The findings suggest the feasibility of integrating OLA into existing treatment regimens, potentially improving clinical outcomes for ALS patients. However, further research must validate these findings in human clinical trials.
Collapse
Affiliation(s)
- Ramaish Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India), Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India), Moga, Punjab, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India), Moga, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India), Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
146
|
Shen LS, Chen JW, Gong RH, Lin Z, Lin YS, Qiao XF, Hu QM, Yang Y, Chen S, Chen GQ. β,β-Dimethylacrylalkannin, a key component of Zicao, induces cell cycle arrest and necrosis in hepatocellular carcinoma cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155959. [PMID: 39178682 DOI: 10.1016/j.phymed.2024.155959] [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: 05/12/2024] [Revised: 08/02/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND β,β-Dimethylacrylalkannin (DMAKN), a natural naphthoquinone found in Zicao, a traditional Chinese medicine (TCM), serves as the designated quantitative marker in the Chinese Pharmacopoeia. Despite its established role in assessing Zicao quality, DMAKN's biological potential remains underexplored in research. METHODS We investigated DMAKN's involvement in Zicao's anti-hepatocellular carcinoma (HCC) properties using a combination of HPLC content analysis and comprehensive bioinformatics. Subsequently, both in vitro and in vivo experiments were conducted to evaluate DMAKN's efficacy against HCC. Mechanistic investigations focused on elucidating DMAKN's impact on cell cycle regulation and induction of cell death. RESULTS Integrated HPLC analysis and bioinformatics identified DMAKN as the primary active compound responsible for Zicao's anti-HCC activity. In vitro and in vivo studies confirmed DMAKN's potent efficacy against HCC. Notably, DMAKN demonstrated dual effects on HCC cells: inhibiting proliferation at lower doses and inducing rapid cell death at higher doses. Mechanistic insights revealed that low-dose DMAKN induced G2/M phase cell cycle arrest through modulation of CDK1 and Cdc25C phosphorylation, while high-dose DMAKN triggered necrosis. Importantly, high-dose DMAKN caused a sharp increase in intracellular ROS levels in a short time, while low-dose DMAKN gradually increased ROS levels over a long period. Additionally, low-dose DMAKN-induced ROS activated the JNK pathway, crucial for cell cycle arrest, whereas high-dose DMAKN-induced necrosis was ROS-dependent but JNK-independent. CONCLUSION This study underscores DMAKN's pivotal role as the principal anti-HCC compound in Zicao, delineating its differential effects and underlying mechanisms. These results demonstrate the potential of DMAKN as a therapeutic agent for the treatment of HCC, providing important information for further study and advancement in cancer therapy.
Collapse
Affiliation(s)
- Li-Sha Shen
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, PR China; Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, PR China
| | - Jia-Wen Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, PR China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, PR China
| | - Rui-Hong Gong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., PR China
| | - Zesi Lin
- Southern Medical University Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Southern Medical University, Guangzhou 510315, PR China
| | - Yu-Shan Lin
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, PR China
| | - Xing-Fang Qiao
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, PR China; Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, PR China
| | - Qian-Mei Hu
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, PR China; Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, PR China
| | - Yong Yang
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, PR China; Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, PR China.
| | - Sibao Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, PR China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, PR China; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., PR China; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., PR China.
| | - Guo-Qing Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, PR China; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., PR China; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., PR China.
| |
Collapse
|
147
|
Li W, Jiang Z, Yan Z, Chen Z, Li L, Wang D, Wang J, Li L, Yang H, Deng J, Lin J. Hydrogel based on M1 macrophage lysate and alginate loading with oxaliplatin for effective immunomodulation to inhibit melanoma progression, recurrence and metastasis. Int J Biol Macromol 2024; 280:135542. [PMID: 39276890 DOI: 10.1016/j.ijbiomac.2024.135542] [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: 06/07/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Despite the monumental success of immunotherapy in treating melanoma clinically, it still confronts significant challenges, chiefly that singular immunomodulatory tactics are insufficient to suppress the recurrence and metastasis of melanoma. Herein, these challenges are addressed by a hydrogel based on M1 macrophage lysate and alginate (M1LMHA) loaded with oxaliplatin (OXA), named M1LMHA@OXA.The results obtained from scanning electron microscopy and confocal microscopy indicate that the structure and morphology of M1LMHA@OXA remain unchanged. Flow cytometry results reveal that M1LMHA@OXA significantly promotes the maturation of dendritic cells (DCs) and enhances the proliferation of T lymphocytes. In a subcutaneous melanoma transplant model, M1LMHA@OXA effectively suppressed tumor growth in comparison to OXA alone and M1LMHA alone. Flow cytometry demonstrated that M1LMHA@OXA markedly increased the number of mature DCs and CD8+ T cells at the tumor site, while significantly reducing the quantity of M2-like tumor-associated macrophages (TAM) and enhancing the presence of M1 macrophages. Enzyme-linked immunosorbent assay (ELISA) results indicated that following treatment with M1LMHA@OXA, the levels of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) in the bloodstream of mice were significantly elevated, whereas interleukin-10 (IL-10) exhibited no significant difference. This outcome further corroborates the ability of M1LMHA@OXA to substantially bolster the immune capability of mice. Similar results have also been observed in a melanoma subcutaneous transplantation recurrence model, and optical imaging of the lungs of mice revealed that M1LMHA@OXA inhibited tumor metastasis to the lungs. Notably, M1LMHA@OXA exhibits an exceptional therapeutic effect on the growth, post-surgical recurrence, and metastasis of the B16F10 melanoma. Therefore, this study provides a straightforward strategy that leverages the cooperative regulation of multiple immune cells to thwart the proliferation, recurrence, and spread of melanoma.
Collapse
Affiliation(s)
- Wanyu Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhonghao Jiang
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhuo Yan
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhihao Chen
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Lianhai Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Dan Wang
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Li Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Huiling Yang
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Junjie Deng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.
| | - Jiantao Lin
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
148
|
Shen F, Zheng YS, Dong L, Cao Z, Cao J. Enhanced tumor suppression in colorectal cancer via berberine-loaded PEG-PLGA nanoparticles. Front Pharmacol 2024; 15:1500731. [PMID: 39555093 PMCID: PMC11563832 DOI: 10.3389/fphar.2024.1500731] [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: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Colorectal cancer (CRC) stands as the third most widespread cancer globally with poor prognosis. Berberine (Ber), as one herbal phytochemical, showed promise in CRC therapy, but its exact mechanism is unclear. Small molecule traditional drugs face challenges in quick metabolism and low bio-availability after systemic administration. Nanodrug deliver system, with their unique properties, has the advantages of protecting drugs, improving drug bio-availability, and reducing toxic and side effects, which exhibited huge drug delivery potential. Herein, the PEG-PLGA nanocarrier was used for encapsulated Ber according to nanoprecipitation and obtained nanomedicine, denoted as NPBer. In vitro, the flow cytometry test and CCK8 assays indicated that NPBer was more easily taken up by HCT116 CRC cells, and had stronger inhibition on cell proliferation with the increase of drug concentration. In addition, RNA-Seq was employed to explore the alterations in the transcriptomes of cancer cells subsequent to treatment with Free Ber or NPBer.The sequencing results indicate that Free Ber could activate cellular aging mechanisms, intensified the iron death pathway, optimized oxidative phosphorylation efficiency, exacerbated apoptosis, accelerated programmed cell death, and negatively modulated key signaling pathways in CRC cells including Wnt, TGF-beta, Hippo, and mTOR signaling pathways. Based on PEG-PLGA nanocarriers, NPBer can improve the in vivo delivery efficiency of Ber, thereby enhancing its antitumor efficacy in vivo, enhancing apoptosis by enhancing the mitochondrial autophagy and autophagy activities of CRC cells, negatively regulating the inflammatory mediator to regulate TRP channels, and inhibiting the activation of Notch signaling pathway. In vivo, NPBer can significantly improve its accumulation and durable drug targeting in tumor site, resulting in induce maximum cell apoptosis and effectively inhibit the proliferation of HCT116 tumor. This strategy provided a promising antitumor therapeutic strategy using Ber-based drugs.
Collapse
Affiliation(s)
- Fei Shen
- Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of General Surgery, Guangzhou Digestive Disease Center, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yun-Sheng Zheng
- Department of General Surgery, Guangzhou Digestive Disease Center, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lan Dong
- Department of General Surgery, Guangzhou Digestive Disease Center, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ziyang Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jie Cao
- Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of General Surgery, Guangzhou Digestive Disease Center, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
149
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [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: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
150
|
Ning N, Li X, Nan Y, Chen G, Huang S, Du Y, Gu Q, Li W, Yuan L. Molecular mechanism of Saikosaponin-d in the treatment of gastric cancer based on network pharmacology and in vitro experimental verification. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8943-8959. [PMID: 38864908 DOI: 10.1007/s00210-024-03214-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
The study aimed to utilize network pharmacology combined with cell experiments to research the mechanism of action of Saikosaponin-d in the treatment of gastric cancer. Drug target genes were obtained from the PubChem database and the Swiss Target Prediction database. Additionally, target genes for gastric cancer were obtained from the GEO database and the Gene Cards database. The core targets were then identified and further analyzed through gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and GESA enrichment. The clinical relevance of the core targets was assessed using the GEPIA and HPA databases. Molecular docking of drug monomers and core target proteins was performed using Auto Duck Tools and Pymol software. Finally, in vitro cellular experiments including cell viability, apoptosis, cell scratch, transwell invasion, transwell migration, qRT-PCR, and Western blot were conducted to verify these findings of network pharmacology. The network pharmacology analysis predicted that the drug monomers interacted with 54 disease targets. Based on clinical relevance analysis, six core targets were selected: VEGFA, IL2, CASP3, BCL2L1, MMP2, and MMP1. Molecular docking results showed binding activity between the Saikosaponin-d monomer and these core targets. Saikosaponin-d could inhibit gastric cancer cell proliferation, induce apoptosis, and inhibit cell migration and invasion.
Collapse
Affiliation(s)
- Na Ning
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xiangyang Li
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Nan
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Guoqing Chen
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Shicong Huang
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Qian Gu
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Weiqiang Li
- Department of Chinese Medical Gastrointestinal, TCM Hospital of Ningxia Medical University, Wuzhong, 751100, China.
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|