1
|
Tran MN, Kim NS, Lee S. Biological network comparison identifies a novel synergistic mechanism of Ginseng Radix-Astragali Radix herb pair in cancer-related fatigue. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118447. [PMID: 38885914 DOI: 10.1016/j.jep.2024.118447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/01/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng Radix and Astragali Radix are commonly combined to tonify Qi and alleviate fatigue. Previous studies have employed biological networks to investigate the mechanisms of herb pairs in treating different diseases. However, these studies have only elucidated a single network for each herb pair, without emphasizing the superiority of the herb combination over individual herbs. AIM OF THE STUDY This study proposes an approach of comparing biological networks to highlight the synergistic effect of the pair in treating cancer-related fatigue (CRF). METHODS The compounds and targets of Ginseng Radix, Astragali Radix, and CRF diseases were collected and predicted using different databases. Subsequently, the overlapping targets between herbs and disease were imported into the STRING and DAVID tools to build protein-protein interaction (PPI) networks and analyze enriched KEGG pathways. The biological networks of Ginseng Radix and Astragali Radix were compared separately or together using the DyNet application. Molecular docking was used to verify the predicted results. Further, in vitro experiments were conducted to validate the synergistic pathways identified in in silico studies. RESULTS In the PPI network comparison, the combination created 89 new interactions and an increased average degree (11.260) when compared to single herbs (10.296 and 9.394). The new interactions concentrated on HRAS, STAT3, JUN, and IL6. The topological analysis identified 20 core targets of the combination, including three Ginseng Radix-specific targets, three Astragali Radix-specific targets, and 14 shared targets. In KEGG enrichment analysis, the combination regulated additional signaling pathways (152) more than Ginseng Radix (146) and Astragali Radix (134) alone. The targets of the herb pair synergistically regulated cancer pathways, specifically hypoxia-inducible factor 1 (HIF-1) signaling pathway. In vitro experiments including enzyme-linked immunosorbent assay and Western blot demonstrated that two herbs combination could up-regulate HIF-1α signaling pathway at different combined concentrations compared to either single herb alone. CONCLUSION The herb pair increased protein interactions and adjusted metabolic pathways more than single herbs. This study provides insights into the combination of Ginseng Radix and Astragali Radix in clinical practice.
Collapse
Affiliation(s)
- Minh Nhat Tran
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea; Korean Convergence Medical Science, University of Science and Technology, Daejeon, Republic of Korea; Faculty of Traditional Medicine, Hue University of Medicine and Pharmacy, Hue University, Thua Thien Hue, Viet Nam.
| | - No Soo Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea.
| | - Sanghun Lee
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea; Korean Convergence Medical Science, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
2
|
Wu W, Song W, Zhao J, Guo S, Hong M, Zheng J, Hua Y, Cao P, Liu R, Duan JA. Saiga antelope horn suppresses febrile seizures in rats by regulating neurotransmitters and the arachidonic acid pathway. Chin Med 2024; 19:78. [PMID: 38831318 PMCID: PMC11149251 DOI: 10.1186/s13020-024-00949-3] [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: 04/11/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Saiga antelope horn (SAH) is a traditional Chinese medicine for treating febrile seizure (FS) with precise efficacy, but its mechanism of action and functional substances are still unclear. Given the need for further research on SAH, our group conducted studies to elucidate its mechanisms and active substances. METHODS An FS rat pup model was constructed through intraperitoneal injection of LPS and hyperthermia induction. Behavioural indicators of seizures, hippocampal histopathological alterations, serum levels of inflammatory cytokines and hippocampal levels of neurotransmitters were observed and measured to investigate the effects of SAH on FS model rats. Hippocampal metabolomics and network pharmacology analyses were conducted to reveal the differential metabolites, key peptides and pathways involved in the suppression of FS by SAH. RESULTS SAH suppressed FS, decreased the inflammatory response and regulated the Glu-GABA balance. Metabolomic analysis revealed 13 biomarkers of FS, of which SAH improved the levels of 8 differential metabolites. Combined with network pharmacology, a "biomarker-core target-key peptide" network was constructed. The peptides of SAH, such as YGQL and LTGGF, could exert therapeutic effects via the arachidonic acid pathway. Molecular docking and ELISA results indicated that functional peptides of SAH could bind to PTGS2 target, inhibiting the generation of AA and its metabolites in hippocampal samples. CONCLUSION In summary, the functional peptides contained in SAH are the main material basis for the treatment of FS, potentially acting through neurotransmitter regulation and the arachidonic acid pathway.
Collapse
Affiliation(s)
- Wenxing Wu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, No 138 Xianlin Road, Nanjing, 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing, 210023, China
| | - Wencong Song
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, No 138 Xianlin Road, Nanjing, 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jingjing Zhao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, No 138 Xianlin Road, Nanjing, 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Sheng Guo
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, No 138 Xianlin Road, Nanjing, 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Min Hong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jie Zheng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yongqing Hua
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Peng Cao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, No 138 Xianlin Road, Nanjing, 210023, China
- Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing, 210023, China
| | - Rui Liu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, No 138 Xianlin Road, Nanjing, 210023, China.
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing, 210023, China.
| | - Jin-Ao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing University of Chinese Medicine, No 138 Xianlin Road, Nanjing, 210023, China.
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Animal-Derived Chinese Medicine and Functional Peptides International Collaboration Joint Laboratory, Nanjing, 210023, China.
| |
Collapse
|
3
|
Sun GF, Qu XH, Jiang LP, Chen ZP, Wang T, Han XJ. The mechanisms of natural products for eye disorders by targeting mitochondrial dysfunction. Front Pharmacol 2024; 15:1270073. [PMID: 38725662 PMCID: PMC11079200 DOI: 10.3389/fphar.2024.1270073] [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: 07/31/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
The human eye is susceptible to various disorders that affect its structure or function, including glaucoma, age-related macular degeneration (AMD) and diabetic retinopathy (DR). Mitochondrial dysfunction has been identified as a critical factor in the pathogenesis and progression of eye disorders, making it a potential therapeutic target in the clinic. Natural products have been used in traditional medicine for centuries and continue to play a significant role in modern drug development and clinical therapeutics. Recently, there has been a surge in research exploring the efficacy of natural products in treating eye disorders and their underlying physiological mechanisms. This review aims to discuss the involvement of mitochondrial dysfunction in eye disorders and summarize the recent advances in the application of natural products targeting mitochondria. In addition, we describe the future perspective and challenges in the development of mitochondria-targeting natural products.
Collapse
Affiliation(s)
- Gui-Feng Sun
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Xin-Hui Qu
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- The Second Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Li-Ping Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Zhi-Ping Chen
- Department of Critical Care Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Tao Wang
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- The Second Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
4
|
Zhou TT, Zhu WJ, Feng H, Ni Y, Li ZW, Sun DD, Li L, Tan JN, Yu CT, Shen WX, Cheng HB. A network pharmacology integrated serum pharmacochemistry strategy for uncovering efficacy of YXC on hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117125. [PMID: 37699493 DOI: 10.1016/j.jep.2023.117125] [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: 05/15/2023] [Revised: 07/28/2023] [Accepted: 09/03/2023] [Indexed: 09/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The YangzhengXiaoji capsule (YXC) has a wide range of applications as effective traditional Chinese medicine (TCM) preparation for hepatocellular carcinoma (HCC) in China. However, the potential bioactive components and the mechanisms are yet unclear. AIM OF THE STUDY The treatment mechanism of YXC on HCC using a network pharmacology integrated serum pharmacochemistry strategy to investigate associated targets and pathways. MATERIALS AND METHODS We utilised HPLC-Q-TOF-MS/MS technology to identify components of the serum samples from both the model group and the YXC (H) group serum, which were collected from nude mice with orthotopic liver tumours. Following this, we conducted compound-target prediction and identified the overlap between the target genes in the YXC group and the oncogenes associated with HCC. The anticancer mechanisms of YXC were investigated by creating a compound-target-pathway network using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) analysis. The anticancer efficacy was evaluated in vitro and in vivo. Also, potential predictive targets and pathways associated with YXC in HCC treatment were assessed by western blotting. RESULTS The YXC (H) serum had 47 bioactive compounds compared to other models, and identified 173 specific target genes. Using the compound-target-disease network, 141 possible target genes were identified. The KEGG pathway analysis revealed vital enrichment of pathways associated with HCC, including regulating Oncology related pathways of inflammation, immunity, apoptosis, and necrosis biological processes. YXC significantly inhibited HCC cell growth in vitro and in vivo. After YXC treatment, western blotting detected alterations in the p53/Bcl-2/Bax/Caspase-3 and PI3K/Akt pathways. CONCLUSIONS YXC can inhibit HCC development and advancement by a variety of components, targets and pathways, especially apoptosis-induction.
Collapse
Affiliation(s)
- Ting-Ting Zhou
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Wen-Jian Zhu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Hui Feng
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Yue Ni
- Yancheng Hospital of Traditional Chinese Medicine, 224000, Yancheng, China
| | - Zi-Wen Li
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Dong-Dong Sun
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China; Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 210023, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumour, 210023, Nanjing, China
| | - Liu Li
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China; Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 210023, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumour, 210023, Nanjing, China
| | - Jia-Ni Tan
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China; Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 210023, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumour, 210023, Nanjing, China
| | - Cheng-Tao Yu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China; Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 210023, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumour, 210023, Nanjing, China
| | - Wei-Xing Shen
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China; Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 210023, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumour, 210023, Nanjing, China.
| | - Hai-Bo Cheng
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, China; Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, 210023, Nanjing, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumour, 210023, Nanjing, China.
| |
Collapse
|
5
|
Li J, Fu R, Guo X, Pan Z, Xie J. Acupuncture improves immunity and fatigue after chemotherapy in breast cancer patients by inhibiting the Leptin/AMPK signaling pathway. Support Care Cancer 2023; 31:506. [PMID: 37542585 PMCID: PMC10404187 DOI: 10.1007/s00520-023-07967-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/24/2023] [Indexed: 08/07/2023]
Abstract
OBJECTIVE Acupuncture has become a popular complementary treatment in oncology. This study is based on RNA-Seq transcriptome sequencing technology to investigate the molecular mechanisms underlying the effect of acupuncture-mediated regulation of the Leptin/AMPK signaling pathway on mitochondrial dysfunction-induced fatigue in breast cancer patients after chemotherapy. METHODS Peripheral blood samples from 10 patients with post-operative chemotherapy for breast cancer were selected for transcriptome sequencing to screen the key molecular pathways involved in fatigue after chemotherapy in breast cancer patients. Besides, peripheral blood samples were collected from 138 post-operative chemotherapy patients with breast cancer to study the composite fatigue and quality of life scores. Flow cytometry was used to detect T lymphocyte subsets in peripheral blood-specific immune cells. In addition, a blood cell analyzer was used to measure peripheral blood leukocyte counts, and MSP-PCR was used to detect mitochondrial DNA mutations in peripheral blood leukocytes. RESULTS Transcriptome bioinformatics analysis screened 147 up-regulated mRNAs and 160 down-regulated mRNAs. Leptin protein was confirmed as the key factor. Leptin was significantly higher in the peripheral blood of breast cancer patients who developed fatigue after chemotherapy. Acupuncture treatment effectively improved post-chemotherapy fatigue and immune status in breast cancer patients, suppressed the expression of Leptin/AMPK signaling pathway-related factor and leukocyte counts, and significantly reduced the rate of mitochondrial DNA mutations in peripheral blood leukocytes. CONCLUSION The Leptin/AMPK signaling pathway may be the key molecular pathway affecting the occurrence of fatigue after chemotherapy in breast cancer patients. Leptin may improve post-chemotherapy fatigue in breast cancer patients by activating AMPK phosphorylation and alleviating mitochondrial functional impairment.
Collapse
Affiliation(s)
- Jinxia Li
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Ruiyang Fu
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Xiaoqing Guo
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Zhongqiang Pan
- Department of Acupuncture, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Huzhou, 313000, Zhejiang, People's Republic of China
| | - Jingjun Xie
- Department of Rehabilitation Medicine, The First People's Hospital of Huzhou, No. 158, Guangchang Hou Road, Huzhou, 313000, Zhejiang, People's Republic of China.
| |
Collapse
|
6
|
Bi Y, Liu X, Liu Y, Wang M, Shan Y, Yin Y, Meng X, Sun F, Li H, Li Z. Molecular and biochemical investigations of the anti-fatigue effects of tea polyphenols and fruit extracts of Lycium ruthenicum Murr. on mice with exercise-induced fatigue. Front Mol Biosci 2023; 10:1223411. [PMID: 37416624 PMCID: PMC10319583 DOI: 10.3389/fmolb.2023.1223411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023] Open
Abstract
Background: The molecular mechanisms regulating the therapeutic effects of plant-based ingredients on the exercise-induced fatigue (EIF) remain unclear. The therapeutic effects of both tea polyphenols (TP) and fruit extracts of Lycium ruthenicum (LR) on mouse model of EIF were investigated. Methods: The variations in the fatigue-related biochemical factors, i.e., lactate dehydrogenase (LDH), superoxide dismutase (SOD), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-2 (IL-2), and interleukin-6 (IL-6), in mouse models of EIF treated with TP and LR were determined. The microRNAs involved in the therapeutic effects of TP and LR on the treatment of mice with EIF were identified using the next-generation sequencing technology. Results: Our results revealed that both TP and LR showed evident anti-inflammatory effect and reduced oxidative stress. In comparison with the control groups, the contents of LDH, TNF-α, IL-6, IL-1β, and IL-2 were significantly decreased and the contents of SOD were significantly increased in the experimental groups treated with either TP or LR. A total of 23 microRNAs (21 upregulated and 2 downregulated) identified for the first time by the high-throughput RNA sequencing were involved in the molecular response to EIF in mice treated with TP and LR. The regulatory functions of these microRNAs in the pathogenesis of EIF in mice were further explored based on Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses with a total of over 20,000-30,000 target genes annotated and 44 metabolic pathways enriched in the experimental groups based on GO and KEGG databases, respectively. Conclusion: Our study revealed the therapeutic effects of TP and LR and identified the microRNAs involved in the molecular mechanisms regulating the EIF in mice, providing strong experimental evidence to support further agricultural development of LR as well as the investigations and applications of TP and LR in the treatment of EIF in humans, including the professional athletes.
Collapse
Affiliation(s)
- Yingxin Bi
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Xianjun Liu
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Yue Liu
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Mengyuan Wang
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Yuhe Yin
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Xianglong Meng
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
| | - Fengjie Sun
- School of Science and Technology, Georgia Gwinnett College, Lawrenceville, GA, United States
| | - Hao Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Zhandong Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| |
Collapse
|
7
|
Hu L, Luo J, Wen G, Sun L, Liu W, Hu H, Li J, Wang L, Su W, Lin L. Identification of the active compounds in the Yi-Fei-San-Jie formula using a comprehensive strategy based on cell extraction/UPLC-MS/MS, network pharmacology, and molecular biology techniques. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154843. [PMID: 37149966 DOI: 10.1016/j.phymed.2023.154843] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND Chinese herbal formulae has multiple active constituents and targets, and the good clinical response is encouraging more scientists to explore the bio-active ingredients in such complex systems. Yi-Fei-San-Jie formula (YFSJF) is commonly used to treat patients with lung cancer in South China; however, its bio-active ingredients remain unknown. PURPOSE We investigated the bio-active ingredients of the YFSJF using a novel comprehensive strategy. METHODS A549 cell extraction coupled with ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS/MS) was used for the screening of potential bio-active ingredients. Network pharmacology approach and molecular dynamics simulation were performed for the screening of targets. Surface plasmon resonance (SPR) assay and molecular biology techniques were used to verify the targets. RESULTS Nine A549 cell membrane-binding compounds were identified through cell extraction/UPLC-MS/MS. Five compounds, namely ginsenoside Ro, ginsenoside Rb1, ginsenoside Rc, peimisine, and peimine were cytotoxic to A549 cells, and they were considered the bio-active ingredients of the YFSJF in vitro. Network pharmacology analysis revealed that TGFBR2 is the key target and the TGFβ pathway is the key pathway targeted by YFSJF in non-small cell lung cancer. Peimisine showed an affinity to TGFBR2 using molecular docking and dynamic stimulation, which was confirmed using surface plasmon resonance spectroscopy. The molecular biology-based analysis further confirmed that peimisine targets TGFBR2 and can reverse A549 epithelial-mesenchymal transition by inhibiting the TGFβ pathway. CONCLUSION Taken together, cell extraction/UPLC-MS/MS, network pharmacology, and molecular biology-based analysis comprise a feasible strategy to explore active ingredients in YFSJF.
Collapse
Affiliation(s)
- Leihao Hu
- School of the First Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510402, China
| | - Jiamin Luo
- School of the First Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Guiqing Wen
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Lingling Sun
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510402, China
| | - Wei Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Hao Hu
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510402, China
| | - Jing Li
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410000, China
| | - Lisheng Wang
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Weiwei Su
- Guangdong Engineering & Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China.
| | - Lizhu Lin
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510402, China.
| |
Collapse
|
8
|
Liu R, Li T, Xu H, Yu G, Zhang T, Wang J, Sun Y, Bi Y, Feng X, Wu H, Zhang C, Sun Y. Systems biology strategy through integrating metabolomics and network pharmacology to reveal the mechanisms of Xiaopi Hewei Capsule improves functional dyspepsia. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1226:123676. [PMID: 37329776 DOI: 10.1016/j.jchromb.2023.123676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/05/2023] [Accepted: 03/13/2023] [Indexed: 06/19/2023]
Abstract
Functional dyspepsia (FD) is one of the more common functional disorders, with a prevalence of 20-25 %. It seriously affects the quality life of patients. Xiaopi Hewei Capsule (XPHC) is a classic formula originated from the Chinese Miao minority. Clinical studies have demonstrated that XPHC can effectively alleviate the symptoms of FD, but the molecular mechanism has not been elucidated. The purpose of this work is to investigate the mechanism of XPHC on FD by integrating metabolomics and network pharmacology. The mice models of FD were established, and gastric emptying rate, small intestine propulsion rate, serum level of motilin and gastrin were evaluate to study the interventional effect of XPHC on FD. Next, a metabolomics strategy has been developed to screen differential metabolites and related metabolic pathways induced by XPHC. Then, prediction of active compounds, targets and pathways of XPHC in treating FD were carried out by commonly used network pharmacological method. Finally, two parts of the results were integrated to investigate therapeutic mechanism of XPHC on FD, which were preliminary validated based on molecular docking. Thus, twenty representative different metabolites and thirteen related pathways of XPHC in treating FD were identified. Most of these metabolites were restored using modulation after XPHC treatment. The results of the network pharmacology analysis showed ten crucial compounds and nine hub genes related to the treatment of FD with XPHC. The further integrated analysis focused on four key targets, such as albumin (ALB), epidermal growth factor receptor (EGFR), tumor necrosis factor (TNF) and roto-oncogene tyrosine-protein kinase Src (SRC), and three representative biomarkers such as citric acid, L-leucine and eicosapentaenoic acid. Furthermore, molecular docking results showed that ten bioactive compounds from XPHC have good binding interactions with the four key genes. The functional enrichment analysis indicated that the potential mechanism of XPHC in treating FD was mainly associated with energy metabolism, amino acid metabolism, lipid metabolism, inflammatory reactions and mucosal repair. Our work confirms that network pharmacology-integrated metabolomics strategyis a powerful means to reveal the therapeutic mechanisms of XPHC improves FD, which contribute its further scientific research.
Collapse
Affiliation(s)
- Runhua Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; Department of Pharmacy, Children's Hospital Affiliated to Capital Institute of Pediatrics, Beijing 100020, China
| | - Tianyi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Haoran Xu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Gengyuan Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tonghua Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaqi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuelin Bi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chenning Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; Department of Pharmacy, Zigong First People's Hospital, Zigong, China.
| | - Yikun Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
9
|
Xiao M, Guo W, Zhang C, Zhu Y, Li Z, Shao C, Jiang J, Yang Z, Zhang J, Lin L. Jian Pi Sheng Sui Gao (JPSSG) alleviation of skeletal myoblast cell apoptosis, oxidative stress, and mitochondrial dysfunction to improve cancer-related fatigue in an AMPK-SIRT1- and HIF-1-dependent manner. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:156. [PMID: 36846003 PMCID: PMC9951005 DOI: 10.21037/atm-22-6611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
Background Jian Pi Sheng Sui Gao (JPSSG), a Chinese traditional herbal paste, possesses certain efficacy in patients with cancer-related fatigue (CRF); however, its related mechanism remains unclear. Hence, network pharmacology analysis, followed by in vivo and in vitro experiments were conducted in this study with the aim to evaluate the effect of JPSSG on CRF and clarify its potential mechanism. Methods Network pharmacology analysis was performed. Subsequently, 12 mice were injected with CT26 cells to establish CRF mouse models and randomly divided into a model group (n=6) and JPSSG group (n=6); meanwhile, another 6 normal mice served as a control group. Then, 3.0 g/kg JPSSG was given to mice in JPSSG group for 15 days, while mice in the n control and model groups received phosphate-buffered saline (PBS) of the same volume for 15 days. For the in vitro experiment, CT26 conditioned medium (CM) was established; meanwhile, the mitochondrial damage model was constructed through C2C12 myotubes stimulated with H2O2. C2C12 myotubes were divided into 5 groups: control group (without treatment), CM group, CM + JPSSG group, H2O2 group, and H2O2 + JGSSP group. Results Network pharmacology analysis identified 87 bioactive compounds and 132 JPSSG-CRF interaction targets. Moreover, according to the Kyoto Encyclopedia of Genes and Genomes enrichment analysis and the subsequent in vivo and in vitro experiments, JPSSG activated adenosine 5'-monophosphate-activated protein kinase-silent-information-regulator factor 2-related-enzyme 1 (AMPK-SIRT1) and hypoxia-inducible factor-1 (HIF-1) signaling pathways during CRF. Moreover, the in vivo experiment showed that JPSSG attenuated CRF in mice, reflected by increased distance traveled, mobile time in open field test, and swimming time in exhaustive swimming test, and decreased absolute rest time and tail suspension test in the JPSSG group (vs. model group). Furthermore, JPSSG upregulated gastrocnemius weight, adenosine triphosphate (ATP), superoxide dismutase (SOD), and the cross-sectional area of the gastrocnemius. With regard to in vitro study, JPSSG elevated cell viability, B-cell lymphoma-2, ATP, SOD, and mitochondrial membrane potential, while it decreased apoptosis rate, cleaved-caspase3, malondialdehyde, and reactive oxygen species in C2C12 myotubes. Conclusions JPSSG ameliorates CRF via alleviating skeletal myoblast cell apoptosis, oxidative stress, and mitochondrial dysfunction in an AMPK-SIRT1- and HIF-1-dependent manner.
Collapse
Affiliation(s)
- Min Xiao
- Clinical Discipline of Integrated Chinese and Western Medicine, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China;,Department of Rheumatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Wei Guo
- Clinical Discipline of Integrated Chinese and Western Medicine, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chi Zhang
- Clinical Discipline of Integrated Chinese and Western Medicine, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yukun Zhu
- Department of Science and Education, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Zhiling Li
- Department of Rheumatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Cui Shao
- Clinical Discipline of Integrated Chinese and Western Medicine, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiling Jiang
- Department of General Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Zhenjiang Yang
- Department of Oncology and Hematology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Jianyong Zhang
- Department of Rheumatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Lizhu Lin
- Clinical Discipline of Integrated Chinese and Western Medicine, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
10
|
Zhou Y, Wu Q, Yu W, Ye F, Cao Y, Akan OD, Wu X, Xie T, Lu H, Cao F, Luo F, Lin Q. Gastrodin ameliorates exercise-induced fatigue via modulating Nrf2 pathway and inhibiting inflammation in mice. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
11
|
Xu H, Xu S, Li L, Wu Y, Mai S, Xie Y, Tan Y, Li A, Xue F, He X, Li Y. Integrated metabolomics, network pharmacology and biological verification to reveal the mechanisms of Nauclea officinalis treatment of LPS-induced acute lung injury. Chin Med 2022; 17:131. [PMID: 36434729 PMCID: PMC9700915 DOI: 10.1186/s13020-022-00685-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a severe inflammatory disease, underscoring the urgent need for novel treatments. Nauclea officinalis Pierre ex Pitard (Danmu in Chinese, DM) is effective in treating inflammatory respiratory diseases. However, there is still no evidence of its protective effect against ALI. METHODS Metabolomics was applied to identify the potential biomarkers and pathways in ALI treated with DM. Further, network pharmacology was introduced to predict the key targets of DM against ALI. Then, the potential pathways and key targets were further verified by immunohistochemistry and western blot assays. RESULTS DM significantly improved lung histopathological characteristics and inflammatory response in LPS-induced ALI. Metabolomics analysis showed that 16 and 19 differential metabolites were identified in plasma and lung tissue, respectively, and most of these metabolites tended to recover after DM treatment. Network pharmacology analysis revealed that the PI3K/Akt pathway may be the main signaling pathway of DM against ALI. The integrated analysis of metabolomics and network pharmacology identified 10 key genes. These genes are closely related to inflammatory response and cell apoptosis of lipopolysaccharide (LPS)-induced ALI in mice. Furthermore, immunohistochemistry and western blot verified that DM could regulate inflammatory response and cell apoptosis by affecting the PI3K/Akt pathway, and expression changes in Bax and Bcl-2 were also triggered. CONCLUSION This study first integrated metabolomics, network pharmacology and biological verification to investigate the potential mechanism of DM in treating ALI, which is related to the regulation of inflammatory response and cell apoptosis. And the integrated analysis can provide new strategies and ideas for the study of traditional Chinese medicines in the treatment of ALI.
Collapse
Affiliation(s)
- Han Xu
- grid.443397.e0000 0004 0368 7493Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Lab of R&D on Tropic Herbs, College of Pharmacy, Hainan Medical University, No. 3 Xueyuan Road, Hainan 571199 Haikou, People’s Republic of China
| | - Sicong Xu
- grid.443397.e0000 0004 0368 7493College of Biomedical Information and Engineering, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, No. 3 Xueyuan Road, Haikou, 571199 Hainan People’s Republic of China
| | - Liyan Li
- grid.443397.e0000 0004 0368 7493Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Lab of R&D on Tropic Herbs, College of Pharmacy, Hainan Medical University, No. 3 Xueyuan Road, Hainan 571199 Haikou, People’s Republic of China
| | - Yuhuang Wu
- grid.443397.e0000 0004 0368 7493Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Lab of R&D on Tropic Herbs, College of Pharmacy, Hainan Medical University, No. 3 Xueyuan Road, Hainan 571199 Haikou, People’s Republic of China
| | - Shiying Mai
- grid.443397.e0000 0004 0368 7493Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Lab of R&D on Tropic Herbs, College of Pharmacy, Hainan Medical University, No. 3 Xueyuan Road, Hainan 571199 Haikou, People’s Republic of China
| | - Yiqiang Xie
- grid.443397.e0000 0004 0368 7493College of Chinese Medicine, Hainan Medical University, No. 3 Xueyuan Road, Haikou, 571199 Hainan People’s Republic of China
| | - Yinfeng Tan
- grid.443397.e0000 0004 0368 7493Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Lab of R&D on Tropic Herbs, College of Pharmacy, Hainan Medical University, No. 3 Xueyuan Road, Hainan 571199 Haikou, People’s Republic of China
| | - Ailing Li
- grid.443397.e0000 0004 0368 7493The Second Affiliated Hospital of Hainan Medical University, 368 Yehai Av., Haikou, 571199 Hainan People’s Republic of China
| | - Fengming Xue
- grid.443397.e0000 0004 0368 7493The Second Affiliated Hospital of Hainan Medical University, 368 Yehai Av., Haikou, 571199 Hainan People’s Republic of China
| | - Xiaoning He
- grid.443397.e0000 0004 0368 7493The Second Affiliated Hospital of Hainan Medical University, 368 Yehai Av., Haikou, 571199 Hainan People’s Republic of China
| | - Yonghui Li
- grid.443397.e0000 0004 0368 7493Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Lab of R&D on Tropic Herbs, College of Pharmacy, Hainan Medical University, No. 3 Xueyuan Road, Hainan 571199 Haikou, People’s Republic of China ,grid.443397.e0000 0004 0368 7493The Second Affiliated Hospital of Hainan Medical University, 368 Yehai Av., Haikou, 571199 Hainan People’s Republic of China
| |
Collapse
|
12
|
Guo W, Yao X, Lan S, Zhang C, Li H, Chen Z, Yu L, Liu G, Lin Y, Liu S, Chen H. Metabolomics and integrated network pharmacology analysis reveal SNKAF decoction suppresses cell proliferation and induced cell apoptisis in hepatocellular carcinoma via PI3K/Akt/P53/FoxO signaling axis. Chin Med 2022; 17:76. [PMID: 35725485 PMCID: PMC9208213 DOI: 10.1186/s13020-022-00628-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 06/02/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND There is no comprehensive treatment method for hepatocellular carcinoma (HCC); hence, research and development are still focused on systemic therapies, including drugs. Sinikangai fang (SNKAF) decoction, a classic Chinese herbal prescription, has been widely used to treat liver cancer. However, there is no research on its core active component and target. METHODS Mouse models were established to measure the anticancer effect of SNKAF decoction on HCC. Further, we investigated the effect of SNKAF decoction on inhibition of hepatoma cells proliferation using cell viability, cloning and invasion assays in vitro. The components of SNKAF were collected from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and TCM@Taiwan database. Metabolomic analysis was used to identify the potential genes and pathways in HCC treated with SNKAF decoction. Then, the expression of phosphoinositide 3-kinase (PI3K), Akt, P53, FoxO proteins of the potential signal pathways were detected using Western blot. RESULTS The animal experiments showed that SNKAF decoction inhibited tumor growth (P < 0.05) and induced no weight loss in the mice. In vitro data showed that HCCLM3 and MHCC97H cell proliferation was inhibited by SNKAF serum in a time- and concentration dependent manner. Further combined analysis network pharmacology with metabonomics showed that 217 target genes overlapped. The core target genes included BCL2, MCL1, Myc, PTEN, gsk3b, CASP9, CREB1, MDM2, pt53 and CCND1. Cancer-associated pathways were largely involved in SNKAF mechanisms, including P53, FoxO, and PI3K/Akt signaling pathways, which are closely related to induced-tumor cell apoptosis. In addition, Western bolt verified that 10% SNKAF serum significantly affected the main proteins of PI3K/Akt/P53/FoxO signaling pathway in both cell lines. CONCLUSION SNKAF decoction-containing serum inhibited HCCLM3 and MHCC97H cell proliferation, migration, invasion, and induced-tumor cell apoptosis in-vivo. We confirmed that SNKAF decoction is a promising alternative treatments for HCC patients.
Collapse
Affiliation(s)
- Wei Guo
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Xiaohui Yao
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Siyuan Lan
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Chi Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Hanhan Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Zhuangzhong Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Ling Yu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Guanxian Liu
- Department of Nephrology, Huizhou Municipal Central Hospital, Huizhou, 510006, Guangdong, People's Republic of China
| | - Yuan Lin
- Department of Pathology, The First Affiliated Hospital of Sun Yat Sen University, Guangzhou, 510080, Guangdong, People's Republic of China.
| | - Shan Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China.
| | - Hanrui Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, People's Republic of China.
| |
Collapse
|
13
|
Zhang Q, Ma W, Wang Q, Gao H, Sun J. Effect of Yang He Decoction on Treatment of Bone Tuberculosis via Phosphoinositide 3-Kinases/Protein Kinase B and Mitogen-Activated Protein Kinase Signaling Pathways. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221075080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Yang He Decoction (YHD), a classical Chinese medicine prescription, is used to treat bone and joint diseases. However, there are few mechanism studies for YHD on the use of YHD to treat bone tuberculosis (BT) and the corresponding mechanism of action of YHD. In the present study, the chemical ingredients of YHD and targets of the ingredients were revealed by a network pharmacology method, and an ingredient–target–disease network was visualized and analyzed. Then, gene ontology enrichment analysis and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were carried out. In addition, molecular docking was performed to clarify the binding of the key active ingredients of YHD to the key targets. Further, an in vitro model of Mycobacterium tuberculosis-induced BT was established, and a mechanism for the inhibitory effect of YHD on the differentiation of RAW 264.7 cells into osteoclasts was investigated. A total of 138 active ingredients in YHD and 50 targets between YHD ingredients and BT were identified. The phosphatidylinositol 3-kinase–protein kinase B (PI3K–Akt) and mitogen-activated protein kinase (MAPK) signaling pathways were the key pathways involved in the anti-BT effect of YHD. Moreover, the in vitro results showed that YHD inhibited the differentiation of RAW 264.7 cells into osteoclasts. YHD decreased the levels of tumor necrosis factor-α and interleukin-1β, increased the levels of superoxide dismutase and glutathione peroxide, and decreased the level of malondialdehyde. Further, YHD inhibited the protein and messenger RNA expression of PI3K/Akt, p38 MAPK, and c-Jun N-terminal kinase. These findings show that YHD is a promising anti-BT agent that suppresses the PI3K/Akt and MAPK signaling pathways to inhibit the differentiation of RAW 264.7 cells into osteoclasts and ameliorate inflammation and oxidative stress.
Collapse
Affiliation(s)
- Qiang Zhang
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
- Shandong Provincial Chest Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Wanli Ma
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Qing Wang
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Hua Gao
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Jianmin Sun
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| |
Collapse
|
14
|
Zhang C, Guo W, Yao X, Xia J, Zhang Z, Li J, Chen H, Lin L. Database mining and animal experiment-based validation of the efficacy and mechanism of Radix Astragali (Huangqi) and Rhizoma Atractylodis Macrocephalae (Baizhu) as core drugs of Traditional Chinese medicine in cancer-related fatigue. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114892. [PMID: 34883219 DOI: 10.1016/j.jep.2021.114892] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/22/2021] [Accepted: 12/01/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In China, Traditional Chinese medicine (TCM) is often used as the main therapy for cancer-related fatigue (CRF). However, there is limited evidence to prove its therapeutic effect and mechanism. AIM OF THE STUDY We aimed to provide a basis for the therapeutic effect of TCM for CRF. MATERIALS AND METHODS We performed a meta-analysis to investigate the efficacy of TCM treatment for CRF. Through frequency statistics and association rule mining, we screened the core Chinese medicine components, Astragalus mongholicus Bunge., root (Radix astragali, Huangqi) and Atractylodes macrocephala Koidz., rhizome (Rhizoma atractylodis macrocephalae, Baizhu). We then used animal experiments to verify the effectiveness of these two TCMs and changes in related indicators in mice. Relevant molecular mechanisms were explored through network pharmacological analysis. RESULTS Twenty-four randomised control trials (RCTs) involving 1865 patients were included in the meta-analysis. TCM produced more positive effects on CRF than standard therapy alone. Radix astragali and Rhizoma atractylodis macrocephalae, as the core drug pair for the treatment of CRF, enhanced the physical fitness of mice; reduced abdominal circumference, level of inflammatory factors, and tumour weight; and increased body weight and blood sugar. Network pharmacology analysis showed that the mechanism of action of Radix astragali and Rhizoma atractylodis macrocephalae on CRF mainly involved compounds, such as quercetin, kaempferol and luteolin, acting through multiple targets, such as Protein kinase B α (AKT1), Tumour necrosis factor (TNF), and Interleukin-6 (IL-6). These molecules regulate cytokines, cancer signalling, and metabolic pathways and confer an anti-CRF effect. CONCLUSIONS TCM may be a promising therapy to relieve CRF in cancer patients. Our research may provide a reference for the clinical application of TCM for treating CRF.
Collapse
Affiliation(s)
- Chi Zhang
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Wei Guo
- The First Hospital Affiliated of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Xiaohui Yao
- The School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Jiangnan Xia
- The School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Zexin Zhang
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jing Li
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Hanrui Chen
- The First Hospital Affiliated of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Lizhu Lin
- The First Hospital Affiliated of Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|