1
|
Abdullah, Ahmad N, Xiao J, Tian W, Khan NU, Hussain M, Ahsan HM, Hamed YS, Zhong H, Guan R. Gingerols: Preparation, encapsulation, and bioactivities focusing gut microbiome modulation and attenuation of disease symptoms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156352. [PMID: 39740381 DOI: 10.1016/j.phymed.2024.156352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 10/17/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Gut dysbiosis, chronic diseases, and microbial recurrent infections concerns have driven the researchers to explore phytochemicals from medicinal and food homologous plants to modulate gut microbiota, mitigate diseases, and inhibit pathogens. Gingerols have attracted attention as therapeutic agents due to their diverse biological activities like gut microbiome regulation, gastro-protective, anti-inflammatory, anti-microbial, and anti-oxidative effects. PURPOSE This review aimed to summarize the gingerols health-promoting potential, specifically focusing on the regulation of gut microbiome, attenuation of disease symptoms, mechanisms of action, and signaling pathways involved. METHOD Research findings from experimental and clinical studies have been summarized regarding gingerols effects on the modulation of gut microbiome and its metabolites, and attenuation of disease symptoms. RESULTS Gingerols are phenolic compounds characterized by a common 3-methoxy-4-hydroxyphenyl moiety in their chemical structures, and further divided into different gingerol types, including gingerols (major), shogaols, paradols, gingerdiols, gingerdiones, and zingerones (minor). Advanced extraction techniques (e.g., ionic liquid-based-, enzyme-assisted-, microwave-assisted-, pressurized liquid-, ultrasound-assisted-, and supercritical fluid extractions) were reported as optimal alternatives to conventional methods for gingerols extraction. Research studies reported that gingerols positively modulated the composition of gut microbiome that helped to combat disease symptoms (e.g., obesity by decreasing weight gain- (Lactobacillus reuteri and Lachnospiraceae) and increasing weight loss associated-bacteria (Akkermansia, Muribaculaceae, and Alloprevotella). Gingerols intervention also ameliorated ulcerative colitis by increasing relative abundance of the beneficial bacteria (Akkermansia, Lachnospiraceae NK4A136, and Muribaculaceae_norank), and decreasing pathogenic microorganisms (Bacteroides, Parabacteroides, and Desulfovibrio). Emerging delivery systems (e.g., microcapsules, nanoparticles, nanostructured lipid carriers, nanoemulsions, and nanoliposomes) can enhance the bioavailability and therapeutic efficacy of gingerols by preserving their inherent properties and addressing challenges of stability, solubility, and absorption. CONCLUSION Gingerols are promising therapeutic agents to modulate gut microbiome (increase beneficial bacteria and inhibit pathogenic microbes), and attenuate chronic disease symptoms such as diabetes, colitis, obesity, oxidative stress, and cancer. Despite significant progress, challenges persist in transforming research findings into industrial applications, such as stability and solubility during processing and low bioavailability in the distal gut to impart desirable health benefits.
Collapse
Affiliation(s)
- Abdullah
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Naveed Ahmad
- Multan College of Food & Nutrition Sciences, Multan Medical & Dental College, Multan, Pakistan
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenni Tian
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Naveed Ullah Khan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Muhammad Hussain
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hafiz Muhammad Ahsan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yahya Saud Hamed
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
2
|
Rutihinda C, Haroun R, Ordonez JP, Mohssine S, Oweida H, Sharma M, Fares M, Ruiz-Dominguez N, Pacheco MFM, Naasri S, Saidi NE, Oweida AJ. Gingerol acts as a potent radiosensitizer in head and neck squamous cell carcinoma. Discov Oncol 2024; 15:553. [PMID: 39397185 PMCID: PMC11471747 DOI: 10.1007/s12672-024-01425-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024] Open
Abstract
Treatment options for advanced head and neck squamous cell carcinoma (HNSCC) are limited and often cause severe toxicity and debilitating long-term impacts. Developing effective and safer treatments is warranted. Several plant extracts have shown their effectiveness, but a comprehensive comparison between plant extracts in HNSCC has not been reported. Our aim was to investigate the effect of different plant extracts on the proliferation and viability of HNSCC cell lines. In addition, we investigated the efficacy of combining cytotoxic plant extracts with radiation. Since RT is a cornerstone in the treatment and management of HNSCC, it is desirable to enhance its efficacy through combination with cytotoxic agents that have minimal side effects. HNSCC cell lines were treated with various plant extracts at different concentrations. MTT assays were performed to identify the most potent anti-tumor plant extract. Colony-formation assays were performed to determine the radiosensitization effect. To investigate the effect on migration, transwell migration assays were performed. Annexin V staining was performed to analyze cell apoptosis. 6-gingerol resulted in the most significant dose-dependent inhibition in all cell lines compared to other plant extracts. Colony-formation assays showed a significant radiosensitizing effect when 6-gingerol was combined with radiation. In addition, the combination of 6-gingerol with radiation resulted in a significant decrease in HNSCC cell migration. Mechanistically, Annexin V staining showed that the combination of 6-gingerol and RT induces a synergistic apoptotic effect in MOC1, MOC2 and SCC9 cells compared to RT alone. In conclusion, 6-gingerol enhances the effect of radiation in HNSCC cell lines and could be a suitable candidate for combination therapy in HNSCC.
Collapse
Affiliation(s)
- Cleopatra Rutihinda
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Ryma Haroun
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Juan Pablo Ordonez
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Saad Mohssine
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Huda Oweida
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Muskaan Sharma
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Mohamed Fares
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Nancy Ruiz-Dominguez
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Maria Fernanda Meza Pacheco
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Sahar Naasri
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Nour Elhouda Saidi
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada
| | - Ayman J Oweida
- Department of Medical Imaging and Radiation Sciences, Faculté de Médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e avenue, Sherbrooke, QC, J1H 5N4, Canada.
| |
Collapse
|
3
|
Sharma P, Gupta K, Khandai SK, Malik S, Thareja S. Phytometabolites as modulators of breast cancer: a comprehensive review of mechanistic insights. Med Oncol 2024; 41:45. [PMID: 38172452 DOI: 10.1007/s12032-023-02269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Breast cancer (BC) is a highly debilitating malignancy affecting females globally and imposing a substantial burden on healthcare systems in both developed and developing nations. Despite the application of conventional therapeutic modalities such as chemotherapy, radiation therapy, and hormonal intervention, BC frequently exhibits resistance, necessitating the urgent development of novel, cost-effective, and accessible treatment strategies. In this context, there is a growing scientific interest in exploring the pharmacological potential of chemical compounds derived from botanical sources, which often exhibit notable biological activity. Extensive in vitro and in vivo investigations have revealed the capacity of these compounds, referred to as phytochemicals, to attenuate the metastatic cascade and reduce the risk of cancer dissemination. These phytochemicals exert their effects through modulation of key molecular and metabolic processes, including regulation of the cell cycle, induction of apoptotic cell death, inhibition of angiogenesis, and suppression of metastatic progression. To shed light on the latest advancements in this field, a comprehensive review of the scientific literature has been conducted, focusing on secondary metabolite agents that have recently been investigated and have demonstrated promising anticancer properties. This review aims to delineate their underlying mechanisms of action and elucidate the associated signaling pathways, thereby contributing to a deeper understanding of their therapeutic potential in the context of BC management.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Khushi Gupta
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sumit Kumar Khandai
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sonia Malik
- Laboratory of Woody Plants and Crops Biology, University of Orleans, Orleans, France
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
4
|
Ni Y, Deng P, Yin R, Zhu Z, Ling C, Ma M, Wang J, Li S, Liu R. Effect and mechanism of paclitaxel loaded on magnetic Fe 3O 4@mSiO 2-NH 2-FA nanocomposites to MCF-7 cells. Drug Deliv 2023; 30:64-82. [PMID: 36474448 PMCID: PMC9744220 DOI: 10.1080/10717544.2022.2154411] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Magnetic Fe3O4 nanoparticles were prepared via a simple hydrothermal method and utilized to load paclitaxel. The average particle size of Fe3O4 nanoparticles was found to be 20.2 ± 3.0 nm, and the calculated saturation magnetization reached 129.38 emu/g, verifying superparamagnetism of nanomaterials. The specific surface area and pore volume were 84.756 m2/g and 0.265 cm3/g, respectively. Subsequently, Fe3O4@mSiO2 nanoparticles were successfully fabricated using the Fe3O4 nanoparticles as precursors with an average size of 27.81 nm. The relevant saturation magnetization, zeta potential, and specific surface area of Fe3O4@mSiO2-NH2-FA were respectively 76.3 emu/g, -14.1 mV, and 324.410 m2/g. The pore volume and average adsorption pore size were 0.369 cm3/g and 4.548 nm, respectively. Compared to free paclitaxel, the solubility and stability of nanoparticles loaded with paclitaxel were improved. The drug loading efficiency and drug load of the nanoformulation were 44.26 and 11.38%, respectively. The Fe3O4@mSiO2-NH2-FA nanocomposites were easy to construct with excellent active targeting performance, pH sensitivity, and sustained-release effect. The nanoformulation also showed good biocompatibility, where the cell viability remained at 73.8% when the concentration reached 1200 μg/mL. The nanoformulation induced cell death through apoptosis, as confirmed by AO/EB staining and flow cytometry. Western blotting results suggested that the nanoformulation could induce iron death by inhibiting Glutathione Peroxidase 4 (GPX4) activity or decreasing Ferritin Heavy Chain 1 (FTH1) expression. Subsequently, the expression of HIF-1α was upregulated owing to the accumulation of reactive oxygen species (ROS), thus affecting the expression of apoptosis-related proteins regulated by p53, inducing cell apoptosis.
Collapse
Affiliation(s)
- Yun Ni
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Peng Deng
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, P.R. China
| | - Ruitong Yin
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Ziye Zhu
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Chen Ling
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Mingyi Ma
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Jie Wang
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China
| | - Shasha Li
- Affiliated Kunshan Hospital, Jiangsu University, Suzhou, P.R. China,CONTACT Shasha Li
| | - Ruijiang Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, P.R. China,Ruijiang Liu
| |
Collapse
|
5
|
Swaminathan H, Saravanamurali K, Yadav SA. Extensive review on breast cancer its etiology, progression, prognostic markers, and treatment. Med Oncol 2023; 40:238. [PMID: 37442848 DOI: 10.1007/s12032-023-02111-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
As the most frequent and vulnerable malignancy among women, breast cancer universally manifests a formidable healthcare challenge. From a biological and molecular perspective, it is a heterogenous disease and is stratified based on the etiological factors driving breast carcinogenesis. Notably, genetic predispositions and epigenetic impacts often constitute the heterogeneity of this disease. Typically, breast cancer is classified intrinsically into histological subtypes in clinical landscapes. These stratifications empower physicians to tailor precise treatments among the spectrum of breast cancer therapeutics. In this pursuit, numerous prognostic algorithms are extensively characterized, drastically changing how breast cancer is portrayed. Therefore, it is a basic requisite to comprehend the multidisciplinary rationales of breast cancer to assist the evolution of novel therapeutic strategies. This review aims at highlighting the molecular and genetic grounds of cancer additionally with therapeutic and phytotherapeutic context. Substantially, it also renders researchers with an insight into the breast cancer cell lines as a model paradigm for breast cancer research interventions.
Collapse
Affiliation(s)
- Harshini Swaminathan
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
| | - K Saravanamurali
- Virus Research and Diagnostics Laboratory, Department of Microbiology, Coimbatore Medical College, Coimbatore, India
| | - Sangilimuthu Alagar Yadav
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India.
| |
Collapse
|
6
|
Chavda VP, Nalla LV, Balar P, Bezbaruah R, Apostolopoulos V, Singla RK, Khadela A, Vora L, Uversky VN. Advanced Phytochemical-Based Nanocarrier Systems for the Treatment of Breast Cancer. Cancers (Basel) 2023; 15:1023. [PMID: 36831369 PMCID: PMC9954440 DOI: 10.3390/cancers15041023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
As the world's most prevalent cancer, breast cancer imposes a significant societal health burden and is among the leading causes of cancer death in women worldwide. Despite the notable improvements in survival in countries with early detection programs, combined with different modes of treatment to eradicate invasive disease, the current chemotherapy regimen faces significant challenges associated with chemotherapy-induced side effects and the development of drug resistance. Therefore, serious concerns regarding current chemotherapeutics are pressuring researchers to develop alternative therapeutics with better efficacy and safety. Due to their extremely biocompatible nature and efficient destruction of cancer cells via numerous mechanisms, phytochemicals have emerged as one of the attractive alternative therapies for chemotherapeutics to treat breast cancer. Additionally, phytofabricated nanocarriers, whether used alone or in conjunction with other loaded phytotherapeutics or chemotherapeutics, showed promising results in treating breast cancer. In the current review, we emphasize the anticancer activity of phytochemical-instigated nanocarriers and phytochemical-loaded nanocarriers against breast cancer both in vitro and in vivo. Since diverse mechanisms are implicated in the anticancer activity of phytochemicals, a strong emphasis is placed on the anticancer pathways underlying their action. Furthermore, we discuss the selective targeted delivery of phytofabricated nanocarriers to cancer cells and consider research gaps, recent developments, and the druggability of phytoceuticals. Combining phytochemical and chemotherapeutic agents with nanotechnology might have far-reaching impacts in the future.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Lakshmi Vineela Nalla
- Department of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Guntur 522302, Andhra Pradesh, India
| | - Pankti Balar
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu 610064, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Avinash Khadela
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Lalitkumar Vora
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33613, USA
| |
Collapse
|
7
|
Zunica ERM, Axelrod CL, Kirwan JP. Phytochemical Targeting of Mitochondria for Breast Cancer Chemoprevention, Therapy, and Sensitization. Int J Mol Sci 2022; 23:ijms232214152. [PMID: 36430632 PMCID: PMC9692881 DOI: 10.3390/ijms232214152] [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: 10/17/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
Breast cancer is a common and deadly disease that causes tremendous physical, emotional, and financial burden on patients and society. Early-stage breast cancer and less aggressive subtypes have promising prognosis for patients, but in aggressive subtypes, and as cancers progress, treatment options and responses diminish, dramatically decreasing survival. Plants are nutritionally rich and biologically diverse organisms containing thousands of metabolites, some of which have chemopreventive, therapeutic, and sensitizing properties, providing a rich source for drug discovery. In this study we review the current landscape of breast cancer with a central focus on the potential role of phytochemicals for treatment, management, and disease prevention. We discuss the relevance of phytochemical targeting of mitochondria for improved anti-breast cancer efficacy. We highlight current applications of phytochemicals and derivative structures that display anti-cancer properties and modulate cancer mitochondria, while describing future applicability and identifying areas of promise.
Collapse
|
8
|
Jiao G, Fan X, Wang Y, Weng N, Ouyang L, Wang H, Pan S, Huang D, Han J, Zhang F, Chen W. Dissection of the Active Ingredients and Potential Mechanism of Han-Shi-Yu-Fei-Decoction in Treating COVID-19 Based on In Vivo Substances Profiling and Clinical Symptom-Guided Network Pharmacology. ACS OMEGA 2022; 7:36598-36610. [PMID: 36268464 PMCID: PMC9578366 DOI: 10.1021/acsomega.2c04589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
This work was aimed to elucidate the mechanism of action of Han-Shi-Yu-Fei-decoction (HSYFD) for treating patients with mild coronavirus disease 2019 (COVID-19) based on clinical symptom-guided network pharmacology. Experimentally, an ultra-high performance liquid chromatography technique coupled with quadrupole time-of-flight mass spectrometry method was used to profile the chemical components and the absorbed prototype constituents in rat serum after its oral administration, and 11 out of 108 compounds were identified. Calculatingly, the disease targets of Han-Shi-Yu-Fei symptoms of COVID-19 were constructed through the TCMIP V2.0 database. The subsequent network pharmacology and molecular docking analysis explored the molecular mechanism of the absorbed prototype constituents in the treatment of COVID-19. A total of 42 HSYFD targets oriented by COVID-19 clinical symptom were obtained, with EGFR, TP53, TNF, JAK2, NR3C1, TH, COMT, and DRD2 as the core targets. Enriched pathway analysis yielded multiple COVID-19-related signaling pathways, such as the PI3K/AKT signaling pathway and JAK-STAT pathway. Molecular docking showed that the key compounds, such as 6-gingerol, 10-gingerol, and scopoletin, had high binding activity to the core targets like COMT, JAK2, and NR3C1. Our work also verified the feasibility of clinical symptom-guided network pharmacology analysis of chemical compounds, and provided a possible agreement between the points of views of traditional Chinese medicine and western medicine on the disease.
Collapse
Affiliation(s)
- Guangyang Jiao
- Institute
of Chinese Materia Medica, Shanghai University
of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiangcheng Fan
- Department
of Pharmacy, Changzheng Hospital, (Second Military Medical University), Naval Medical University, Shanghai 200003, China
- Shanghai
Key Laboratory for Pharmaceutical Metabolite Research, Shanghai 200433, China
| | - Yejian Wang
- Department
of Pharmacology, Anhui University of Chinese
Medicine, Hefei 230012, Anhui, China
| | - Nan Weng
- Department
of Pharmacy, Changzheng Hospital, (Second Military Medical University), Naval Medical University, Shanghai 200003, China
- School
of Traditional Chinese Material, Shenyang
Pharmaceutical University, Shenyang 11001, China
| | - Luolan Ouyang
- School of
Pharmacy, Shanghai University of Chinese
Medicine, Shanghai 201203, China
| | - Haoqian Wang
- School of
Pharmacy, Shanghai University of Chinese
Medicine, Shanghai 201203, China
| | - Sihan Pan
- School of
Pharmacy, Shanghai University of Chinese
Medicine, Shanghai 201203, China
| | - Doudou Huang
- Institute
of Chinese Materia Medica, Shanghai University
of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jun Han
- Department
of Gastroenterology, Changzheng Hospital, (Second Military Medical
University), Naval Medical University, Shanghai 200003, China
| | - Feng Zhang
- Department
of Pharmacy, Changzheng Hospital, (Second Military Medical University), Naval Medical University, Shanghai 200003, China
- Shanghai
Key Laboratory for Pharmaceutical Metabolite Research, Shanghai 200433, China
- Department
of Pharmacology, Anhui University of Chinese
Medicine, Hefei 230012, Anhui, China
| | - Wansheng Chen
- Institute
of Chinese Materia Medica, Shanghai University
of Traditional Chinese Medicine, Shanghai 201203, China
- Department
of Pharmacy, Changzheng Hospital, (Second Military Medical University), Naval Medical University, Shanghai 200003, China
- Shanghai
Key Laboratory for Pharmaceutical Metabolite Research, Shanghai 200433, China
| |
Collapse
|
9
|
Han X, Yang Y, Qi J, Zhang M, Xue Y, Chu X, Jia Q, Sun S, Guan S. Protective effects and possible mechanism of 6-gingerol against arsenic trioxide-induced nephrotoxicity based on network pharmacological analysis and experimental validation. Int Immunopharmacol 2022; 110:108926. [PMID: 35728306 DOI: 10.1016/j.intimp.2022.108926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Nephrotoxicity induced by the chemotherapeutic drug arsenic trioxide (ATO) is often overlooked, and the underlying mechanisms remain poorly understood. Based on network pharmacology and experimental validation, this study investigates the protection of 6-gingerol (6G) against ATO-induced nephrotoxicity and the potential mechanisms. METHODS We screened and collected 6G and disease-related targets and then imported the interaction targets into a String database to construct protein-protein interaction (PPI) networks. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID). Mice were injected intraperitoneally with ATO (5 mg/kg) for seven days to induce nephrotoxicity, and then the histological morphology of the kidneys, biochemical indices of serum and tissues, and associated protein expressions were observed. RESULTS The network pharmacology results revealed that the effects of 6G against nephrotoxicity are closely related to apoptosis, and the MAPKs pathway was screened for validation. In animal experiments, 6G improved the histopathological morphology of the kidneys, reduced the levels of renal function markers, enhanced antioxidant activity, and decreased the levels of inflammation. Furthermore, 6G reduced apoptotic cells in kidney tissues, decreased the levels of Bax and c-Caspase-3, and increased the level of Bcl-2. The results of immunohistochemistry and western blotting revealed that 6G significantly inhibited the expressions of p-p38, p-ERK, and p-JNK. CONCLUSION The results comprehensively demonstrate the protective effects of 6G against ATO-induced nephrotoxicity. The effects are related to anti-oxidant, anti-inflammatory, and anti-apoptotic properties, possibly through inhibition of the MAPKs pathway.
Collapse
Affiliation(s)
- Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yakun Yang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jiaying Qi
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Muqing Zhang
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China; Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yucong Xue
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Xi Chu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qingzhong Jia
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Shijiang Sun
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China.
| | - Shengjiang Guan
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China; School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China.
| |
Collapse
|