1
|
Jin M, Wei L, Wang J, Shen Y, Gao L, Zhao F, Gao Q, Ma Y, Sun Y, Lin Y, Ji G, Cai P, Yan R. Formononetin: a review of its source, pharmacology, drug combination, toxicity, derivatives, and drug delivery systems. Front Pharmacol 2025; 16:1534798. [PMID: 40098623 PMCID: PMC11911920 DOI: 10.3389/fphar.2025.1534798] [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: 11/26/2024] [Accepted: 01/31/2025] [Indexed: 03/19/2025] Open
Abstract
Formononetin (FMN) is a common natural metabolite that can be extracted and isolated from some common botanical drugs. In recent years, FMN has garnered increasing attention due to its beneficial biological activities. In this paper, we systematically summarize the sources of FMN and provide a comprehensive review of its pharmacological activities and molecular mechanisms, co-administration, toxicity, derivatives, and drug delivery systems in the last 5 years. The study results found that FMN has a wide range of pharmacological activities in neurological disorders, organ damage and cancer, showing great potential for clinical application and broad prospects. Researchers are exploring various types of delivery systems, including nanoparticle carriers, ligand modifications and polymer microspheres. These advanced delivery systems can enhance the stability of FMN, prolong its release time in vivo, and improve targeting, thereby optimizing its therapeutic efficacy and reducing side effects, and greatly improving its bioavailability. In conclusion, FMN is a natural metabolite with considerable research value, and its diverse biological activities make it a promising candidate for drug development and medical research.
Collapse
Affiliation(s)
- Min Jin
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linfang Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianhua Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuehong Shen
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Gao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fan Zhao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qianying Gao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yifei Ma
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yongyan Sun
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Lin
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guanjie Ji
- Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Pingping Cai
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rugen Yan
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
Ul Hassan MH, Shahbaz M, Imran M, Momal U, Naeem H, Mujtaba A, Hussain M, Anwar MJ, Alsagaby SA, Al Abdulmonem W, Yehuala TF, Abdelgawad MA, El‐Ghorab AH, Selim S, Mostafa EM. Isoflavones: Promising Natural Agent for Cancer Prevention and Treatment. Food Sci Nutr 2025; 13:e70091. [PMID: 40078339 PMCID: PMC11896816 DOI: 10.1002/fsn3.70091] [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: 10/08/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Isoflavones are currently being investigated by researchers in order to demonstrate their ability to prevent the proliferation of cancer cells. The current review aimed to demonstrate the potential of isoflavones to eliminate cancerous cells in the stomach, liver, lung, breast, and prostate, as their anticancer properties are due to the ability to block the signaling pathways of the extracellular signal-controlled kinase (MAPK/ERK) and proteasome (PI3K/AKT/mTOR). Isoflavones can inhibit the cell division of various cancer cells. Isoflavones can block the androgen receptor (AR), a protein that is required for the growth and dissemination of prostate cancer. It initiates the caspase cascade and obstructs the production of new proteins to eliminate lung cancer cells. These inhibit colon cancer cells by entering their G2/M cell cycle phase and inducing apoptosis. These are also known to inhibit the production of cyclin-dependent kinase 2 and cyclin B1, two proteins that are related to an enhanced risk of colon cancer. These suppress the breakdown of cyclin B1 and CDK2 to stop the development of cancer. Preclinical evidence consistently supports the efficacy of isoflavones in suppressing tumor growth; however, human clinical trials show variability due to differences in bioavailability, metabolism, and dosage. Despite their promise as alternative or adjunctive cancer therapies, limitations such as low solubility, interindividual metabolic variations, and inconsistent clinical outcomes necessitate further large-scale, controlled trials. Future research should focus on improving bioavailability and exploring synergistic effects with conventional therapies.
Collapse
Affiliation(s)
- Muhammad Hammad Ul Hassan
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Muhammad Shahbaz
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Muhammad Imran
- Department of Food Science and TechnologyUniversity of NarowalNarowalPakistan
| | - Ushna Momal
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Hammad Naeem
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
- Post Harvest Research CentreAyub Agricultural Research InstituteFaisalabadPakistan
| | - Ahmed Mujtaba
- Department of Food Science and Technology, Faculty of Engineering Sciences and TechnologyHamdard University Islamabad CampusIslamabadPakistan
| | - Muzzamal Hussain
- Department of Food ScienceGovernment College University FaisalabadFaisalabadPakistan
| | - Muhammad Junaid Anwar
- Department of Food Science and Technology, Faculty of Food Science and NutritionBahauddin Zakariya UniversityMultanPakistan
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesMajmaah UniversityAl‐MajmaahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of MedicineQassim UniversityBuraidahSaudi Arabia
| | - Tadesse Fenta Yehuala
- Faculty of Chemical and Food Engineering, Bahir Dar Institute of TechnologyBahir Dar UniversityBahir Dar CityEthiopia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of PharmacyJouf UniversitySakakaAljoufSaudi Arabia
| | - Ahmed H. El‐Ghorab
- Department of Chemistry, College of ScienceJouf UniversitySakakaSaudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesJouf UniversitySakakaSaudi Arabia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of PharmacyJouf UniversitySakakaSaudi Arabia
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys)Al‐Azhar UniversityCairoEgypt
| |
Collapse
|
3
|
Selepe MA. Isoflavone Derivatives as Potential Anticancer Agents: Synthesis and Bioactivity Studies. ChemMedChem 2024; 19:e202400420. [PMID: 39091268 PMCID: PMC11617652 DOI: 10.1002/cmdc.202400420] [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: 05/31/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/04/2024]
Abstract
Isoflavones are phenolic natural compounds with a C6C3C6 framework. They possess a plethora of biological activities that are associated with putative benefits to human health. In particular, the cancer chemopreventive and chemotherapeutic potential of isoflavones has attracted the interest of researchers. Several isoflavone derivatives have been synthesised and probed for their anticancer activities. The isoflavone analogues are mainly synthesised by molecular hybridisation and other strategies that enable diversification through early or late-stage functionalisation of A-, B- and C-rings of the isoflavones. This has resulted in the discovery of isoflavone analogues with improved antiproliferative activities against several cancer cells and different mechanisms of action. In this review, the synthesis of isoflavone derivatives and their anticancer activity studies are discussed.
Collapse
Affiliation(s)
- Mamoalosi A. Selepe
- Department of Chemistry, Faculty of Natural and Agricultural SciencesUniversity of PretoriaPrivate bag X 20Hatfield0028South Africa
| |
Collapse
|
4
|
Cai J, Tan X, Hu Q, Pan H, Zhao M, Guo C, Zeng J, Ma X, Zhao Y. Flavonoids and Gastric Cancer Therapy: From Signaling Pathway to Therapeutic Significance. Drug Des Devel Ther 2024; 18:3233-3253. [PMID: 39081701 PMCID: PMC11287762 DOI: 10.2147/dddt.s466470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Gastric cancer (GC) is a prevalent gastrointestinal tumor characterized by high mortality and recurrence rates. Current treatments often have limitations, prompting researchers to explore novel anti-tumor substances and develop new drugs. Flavonoids, natural compounds with diverse biological activities, are gaining increasing attention in this regard. We searched from PubMed, Web of Science, SpringerLink and other databases to find the relevant literature in the last two decades. Using "gastric cancer", "stomach cancers", "flavonoid", "bioflavonoid", "2-Phenyl-Chromene" as keywords, were searched, then analyzed and summarized the mechanism of flavonoids in the treatment of GC. It was revealed that the anti-tumor mechanism of flavonoids involves inhibiting tumor growth, proliferation, invasion, and metastasis, as well as inducing cell death through various processes such as apoptosis, autophagy, ferroptosis, and pyroptosis. Additionally, combining flavonoids with other chemotherapeutic agents like 5-FU and platinum compounds can potentially reduce chemoresistance. Flavonoids have also demonstrated enhanced biological activity when used in combination with other natural products. Consequently, this review proposes innovative perspectives for the development of flavonoids as new anti-GC agents.
Collapse
Affiliation(s)
- Jiaying Cai
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiyue Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Maoyuan Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Cui Guo
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Jinhao Zeng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
5
|
Koley M, Han J, Soloshonok VA, Mojumder S, Javahershenas R, Makarem A. Latest developments in coumarin-based anticancer agents: mechanism of action and structure-activity relationship studies. RSC Med Chem 2024; 15:10-54. [PMID: 38283214 PMCID: PMC10809357 DOI: 10.1039/d3md00511a] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/20/2023] [Indexed: 01/30/2024] Open
Abstract
Many researchers around the world are working on the development of novel anticancer drugs with different mechanisms of action. In this case, coumarin is a highly promising pharmacophore for the development of novel anticancer drugs. Besides, the hybridization of this moiety with other anticancer pharmacophores has emerged as a potent breakthrough in the treatment of cancer to decrease its side effects and increase its efficiency. This review aims to provide a comprehensive overview of the recent development of coumarin derivatives and their application as novel anticancer drugs. Herein, we highlight and describe the largest number of research works reported in this field from 2015 to August 2023, along with their mechanisms of action and structure-activity relationship studies, making this review different from the other review articles published on this topic to date.
Collapse
Affiliation(s)
- Manankar Koley
- CSIR-Central Glass & Ceramic Research Institute Kolkata India
| | - Jianlin Han
- College of Chemical Engineering, Nanjing Forestry University Nanjing China
| | - Vadim A Soloshonok
- Department of Organic Chemistry I, University of the Basque Country San Sebastián Spain
- IKERBASQUE, Basque Foundation for Science Bilbao Spain
| | | | - Ramin Javahershenas
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University Urmia Iran
| | - Ata Makarem
- Institute of Pharmacy, University of Hamburg Hamburg Germany
| |
Collapse
|
6
|
Aliya S, Alhammadi M, Park U, Tiwari JN, Lee JH, Han YK, Huh YS. The potential role of formononetin in cancer treatment: An updated review. Biomed Pharmacother 2023; 168:115811. [PMID: 37922652 DOI: 10.1016/j.biopha.2023.115811] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/21/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Currently, cancer is one of the main research topics, due to its high incidence and drug resistance to existing anti-cancer drugs. Formononetin, a natural product with phytoestrogenic properties and diverse biological functions, has attracted the attention of researchers working on anticancer drugs. Formononetin emerges as an intriguing bioactive substance compared to other isoflavones as it exhibits potent chemotherapeutic activity with less toxicity. Formononetin effectively plays a significant role in inhibiting cell proliferation, invasion, and metastatic abilities of cancer cells by targeting major signaling pathways at the junction of interconnected pathways. It also induces apoptosis and cell cycle arrest by modulating mediator proteins. It causes upregulation of key factors such as p-AKT, p38, p21, and p53 and downregulation of NF-κB. Furthermore, formononetin regulates the neoplastic microenvironment by inactivating the ERK1/2 pathway and lamin A/C signaling and has been reported to inactivate JAK/STAT, PKB or AKT, and mitogen-activated protein kinase pathways and to suppress cell migration, invasion, and angiogenesis in human cancer cells. To assist researchers in further exploring formononetin as a potential anticancer therapeutic candidate, this review focuses on both in vitro and in vivo proof of concept studies, patents, and clinical trials pertinent to formononetin's anticancer properties. Overall, this review discusses formononetin from a comprehensive perspective to highlight its potential benefits as an anticancer agent.
Collapse
Affiliation(s)
- Sheik Aliya
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Munirah Alhammadi
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Uichang Park
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Jitendra N Tiwari
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 100-715, Republic of Korea
| | - Jeong-Hwan Lee
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea; Department of Materials Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 100-715, Republic of Korea.
| | - Yun Suk Huh
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
7
|
Zhao W, Zheng XD, Tang PYZ, Li HM, Liu X, Zhong JJ, Tang YJ. Advances of antitumor drug discovery in traditional Chinese medicine and natural active products by using multi-active components combination. Med Res Rev 2023; 43:1778-1808. [PMID: 37183170 DOI: 10.1002/med.21963] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/03/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023]
Abstract
The antitumor efficacy of Chinese herbal medicines has been widely recognized. Leading compounds such as sterols, glycosides, flavonoids, alkaloids, terpenoids, phenylpropanoids, and polyketides constitute their complex active components. The antitumor monomers derived from Chinese medicine possess an attractive anticancer activity. However, their use was limited by low bioavailability, significant toxicity, and side effects, hindering their clinical applications. Recently, new chemical entities have been designed and synthesized by combining natural drugs with other small drug molecules or active moieties to improve the antitumor activity and selectivity, and reduce side effects. Such a novel conjugated drug that can interact with several vital biological targets in cells may have a more significant or synergistic anticancer activity than a single-molecule drug. In addition, antitumor conjugates could be obtained by combining pharmacophores containing two or more known drugs or leading compounds. Based on these studies, the new drug research and development could be greatly shortened. This study reviews the research progress of conjugates with antitumor activity based on Chinese herbal medicine. It is expected to serve as a valuable reference to antitumor drug research and clinical application of traditional Chinese medicine.
Collapse
Affiliation(s)
- Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiao-Di Zheng
- Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | | | - Hong-Mei Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xue Liu
- Jinan Intellectual Property Protection Center, Jinan, China
| | - Jian-Jiang Zhong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
8
|
Wang P, Wang Z, Zhang Z, Cao H, Kong L, Ma W, Ren W. A review of the botany, phytochemistry, traditional uses, pharmacology, toxicology, and quality control of the Astragalus memeranaceus. Front Pharmacol 2023; 14:1242318. [PMID: 37680711 PMCID: PMC10482111 DOI: 10.3389/fphar.2023.1242318] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
Astragali Radix (Huangqi) is mainly distributed in the Northern Hemisphere, South America, and Africa and rarely in North America and Oceania. It has long been used as an ethnomedicine in the Russian Federation, Mongolia, Korea, Kazakhstan, and China. It was first recorded in the Shennong Ben Cao Jing and includes the effects of reinforcing healthy qi, dispelling pathogenic factors, promoting diuresis, reducing swelling, activating blood circulation, and dredging collaterals. This review systematically summarizes the botanical characteristics, phytochemistry, traditional uses, pharmacology, and toxicology of Astragalus to explore the potential of Huangqi and expand its applications. Data were obtained from databases such as PubMed, CNKI, Wan Fang Data, Baidu Scholar, and Google Scholar. The collected material also includes classic works of Chinese herbal medicine, Chinese Pharmacopoeia, Chinese Medicine Dictionary, and PhD and Master's theses. The pharmacological effects of the isoflavone fraction in Huangqi have been studied extensively; The pharmacological effects of Huangqi isoflavone are mainly reflected in its anti-inflammatory, anti-tumor, anti-oxidant, anti-allergic, and anti-diabetic properties and its ability to treat several related diseases. Additionally, the medicinal uses, chemical composition, pharmacological activity, toxicology, and quality control of Huangqi require further elucidation. Here, we provide a comprehensive review of the botany, phytochemistry, traditional uses, pharmacology, toxicology, and quality control of Astragalus to assist future innovative research and to identify and develop new drugs involving Huangqi.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Ma
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Weichao Ren
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
9
|
Nigam M, Mishra AP, Deb VK, Dimri DB, Tiwari V, Bungau SG, Bungau AF, Radu AF. Evaluation of the association of chronic inflammation and cancer: Insights and implications. Biomed Pharmacother 2023; 164:115015. [PMID: 37321055 DOI: 10.1016/j.biopha.2023.115015] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/02/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023] Open
Abstract
Among the most extensively researched processes in the development and treatment of cancer is inflammatory condition. Although acute inflammation is essential for the wound healing and reconstruction of tissues that have been damaged, chronic inflammation may contribute to the onset and growth of a number of diseases, including cancer. By disrupting the signaling processes of cells, which result in cancer induction, invasion, and development, a variety of inflammatory molecules are linked to the development of cancer. The microenvironment surrounding the tumor is greatly influenced by inflammatory cells and their subsequent secretions, which also contribute significantly to the tumor's growth, survivability, and potential migration. These inflammatory variables have been mentioned in several publications as prospective diagnostic tools for anticipating the onset of cancer. Targeting inflammation with various therapies can reduce the inflammatory response and potentially limit or block the proliferation of cancer cells. The scientific medical literature from the past three decades has been studied to determine how inflammatory chemicals and cell signaling pathways related to cancer invasion and metastasis are related. The current narrative review updates the relevant literature while highlighting the specifics of inflammatory signaling pathways in cancer and their possible therapeutic possibilities.
Collapse
Affiliation(s)
- Manisha Nigam
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, 246174 Srinagar Garhwal, Uttarakhand, India
| | - Abhay Prakash Mishra
- Department of Pharmacology, Faculty of Health Science, University of Free State, 9300 Bloemfontein, South Africa.
| | - Vishal Kumar Deb
- Dietetics and Nutrition Technology Division, CSIR Institute of Himalayan Bioresource Technology, 176061 Palampur, Himanchal Pradesh, India
| | - Deen Bandhu Dimri
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, 246174 Srinagar Garhwal, Uttarakhand, India
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi 221005, Uttar Pradesh, India
| | - Simona Gabriela Bungau
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| | - Alexa Florina Bungau
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Andrei-Flavius Radu
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
10
|
Almatroodi SA, Almatroudi A, Khan AA, Rahmani AH. Potential Therapeutic Targets of Formononetin, a Type of Methoxylated Isoflavone, and Its Role in Cancer Therapy through the Modulation of Signal Transduction Pathways. Int J Mol Sci 2023; 24:ijms24119719. [PMID: 37298670 DOI: 10.3390/ijms24119719] [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: 05/07/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Cancer is one of the main causes of death in all developed and developing countries. Various factors are involved in cancer development and progression, including inflammation and alterations in cellular processes and signaling transduction pathways. Natural compounds have shown health-promoting effects through their antioxidant and anti-inflammatory potential, having an important role in the inhibition of cancer growth. In this regard, formononetin, a type of isoflavone, plays a significant role in disease management through the modulation of inflammation, angiogenesis, cell cycle, and apoptosis. Furthermore, its role in cancer management has been proven through the regulation of different signal transduction pathways, such as the signal transducer and activator of transcription 3 (STAT 3), Phosphatidyl inositol 3 kinase/protein kinase B (PI3K/Akt), and mitogen activating protein kinase (MAPK) signaling pathways. The anticancer potential of formononetin has been reported against various cancer types, such as breast, cervical, head and neck, colon, and ovarian cancers. This review focuses on the role of formononetin in different cancer types through the modulation of various cell signaling pathways. Moreover, synergistic effect with anticancer drugs and methods to improve bioavailability are explained. Thus, detailed studies based on clinical trials are required to explore the potential role of formononetin in cancer prevention and treatment.
Collapse
Affiliation(s)
- Saleh A Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia
| |
Collapse
|
11
|
Recent Advances in Natural Product-Based Hybrids as Anti-Cancer Agents. Molecules 2022; 27:molecules27196632. [PMID: 36235168 PMCID: PMC9572494 DOI: 10.3390/molecules27196632] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/24/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is one of the top leading causes of death worldwide. It is a heterogenous disease characterized by unregulated cell proliferation and invasiveness of abnormal cells. For the treatment of cancer, natural products have been widely used as a source of therapeutic ingredients since ancient times. Although natural compounds and their derivatives have demonstrated strong antitumor activity in many types of cancer, their poor pharmacokinetic properties, low cell selectivity, limited bioavailability and restricted efficacy against drug-resistant cancer cells hinder their wide clinical application. Conjugation of natural products with other bioactive molecules has given rise to a new field in drug discovery resulting to the development of novel, bifunctional and more potent drugs for cancer therapy to overcome the current drawbacks. This review discusses multiple categories of such bifunctional conjugates and highlights recent trends and advances in the development of natural product hybrids. Among them, ADCs, PDCs, ApDCs, PROTACs and AUTOTACs represent emerging therapeutic agents against cancer.
Collapse
|
12
|
Ma X, Wang J. Formononetin: A Pathway to Protect Neurons. Front Integr Neurosci 2022; 16:908378. [PMID: 35910340 PMCID: PMC9326316 DOI: 10.3389/fnint.2022.908378] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/14/2022] [Indexed: 11/19/2022] Open
Abstract
Formononetin (FMN) is a phytoestrogen member of the flavonoid family, which has the pharmacological effects of antioxidative, antihypertensive, antitumor, and anti-infective. FMN demonstrates potential in the prevention and treatment of diseases, specifically neurological diseases, such as traumatic brain injury (TBI), spinal cord injury (SCI), ischemic stroke, cerebral ischemia-reperfusion, Alzheimer’s disease, and nerve tumor. Herein, a literature search is conducted to provide information on the signaling pathways of neuroprotection of formononetin based on the neuroprotective study. The significant neuroprotective function of FMN makes it a novel candidate for the development of drugs targeting the central nervous system.
Collapse
Affiliation(s)
- Xiaoyu Ma
- The Second Clinical Medical School, Nanjing Medical University, Nanjing, China
| | - Juejin Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
- *Correspondence: Juejin Wang,
| |
Collapse
|
13
|
Hashem S, Ali TA, Akhtar S, Nisar S, Sageena G, Ali S, Al-Mannai S, Therachiyil L, Mir R, Elfaki I, Mir MM, Jamal F, Masoodi T, Uddin S, Singh M, Haris M, Macha M, Bhat AA. Targeting cancer signaling pathways by natural products: Exploring promising anti-cancer agents. Biomed Pharmacother 2022; 150:113054. [PMID: 35658225 DOI: 10.1016/j.biopha.2022.113054] [Citation(s) in RCA: 145] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is one of the leading causes of death and significantly burdens the healthcare system. Due to its prevalence, there is undoubtedly an unmet need to discover novel anticancer drugs. The use of natural products as anticancer agents is an acceptable therapeutic approach due to accessibility, applicability, and reduced cytotoxicity. Natural products have been an incomparable source of anticancer drugs in the modern era of drug discovery. Along with their derivatives and analogs, natural products play a major role in cancer treatment by modulating the cancer microenvironment and different signaling pathways. These compounds are effective against several signaling pathways, mainly cell death pathways (apoptosis and autophagy) and embryonic developmental pathways (Notch pathway, Wnt pathway, and Hedgehog pathway). The historical record of natural products is strong, but there is a need to investigate the current role of natural products in the discovery and development of cancer drugs and determine the possibility of natural products being an important source of future therapeutic agents. Many target-specific anticancer drugs failed to provide successful results, which accounts for a need to investigate natural products with multi-target characteristics to achieve better outcomes. The potential of natural products to be promising novel compounds for cancer treatment makes them an important area of research. This review explores the significance of natural products in inhibiting the various signaling pathways that serve as drivers of carcinogenesis and thus pave the way for developing and discovering anticancer drugs.
Collapse
Affiliation(s)
- Sheema Hashem
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
| | - Tayyiba Akbar Ali
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
| | - Sabah Akhtar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
| | - Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
| | | | - Shahid Ali
- International Potato Center (CIP), Shillong, Meghalaya, India
| | - Sharefa Al-Mannai
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar
| | - Lubna Therachiyil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha, Qatar
| | - Rashid Mir
- Prince Fahd Bin Sultan Research chair, Department Of Medical Lab Technology, FAMS, University of Tabuk,Saudi Arabia
| | - Imadeldin Elfaki
- Department of Biochemistry, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammad Muzaffar Mir
- Department of Basic Medical Sciences, College of Medicine, University of Bisha, Saudi Arabia
| | - Farrukh Jamal
- Dr. Rammanohar Lohia Avadh University, Ayodhya, India
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Mayank Singh
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar; Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Muzafar Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Kashmir, India.
| | - Ajaz A Bhat
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
14
|
Huang J, Chen X, Xie A. Formononetin ameliorates IL‑13‑induced inflammation and mucus formation in human nasal epithelial cells by activating the SIRT1/Nrf2 signaling pathway. Mol Med Rep 2021; 24:832. [PMID: 34590155 PMCID: PMC8503736 DOI: 10.3892/mmr.2021.12472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/28/2021] [Indexed: 01/18/2023] Open
Abstract
Formononetin has proven to be anti‑inflammatory and able to alleviate symptoms of certain allergic diseases. The present study aimed to determine and elucidate the potential effects of formononetin in allergic rhinitis. JME/CF15 cells were pretreated with formononetin at different doses, followed by stimulation with IL‑13. Cell Counting Kit‑8 assay was performed to determine the cytotoxicity of formononetin. The expression levels of inflammation‑related proteins, histamine, IgE, TNF‑α, IL‑1β, IL‑6, granulocyte‑macrophage colony‑stimulating factor and eotaxin in IL‑13‑stimulated JME/CF15 cells were detected using ELISAs. The expression levels of phosphorylated‑NF‑κB p65, NF‑κB p65 and cyclooxygenase‑2 (Cox‑2) were analyzed using western blotting. Reverse transcription‑quantitative PCR, western blotting and immunofluorescence were performed to measure the levels of mucin 5AC oligomeric mucus/gel‑forming. Expression levels of sirtuin 1 (SIRT1) and nuclear erythroid factor 2‑related factor 2 (Nrf2) proteins were also measured using western blotting. The results of the present study revealed that formononetin exerted no cytotoxic effect on the viability of JME/CF15 cells. Following stimulation of JME/CF15 cells with IL‑13, formononetin suppressed the upregulated expression levels of proinflammatory cytokines. IL‑13‑induced formation of mucus was also attenuated by formononetin treatment. Furthermore, it was found that the SIRT1/Nrf2 signaling pathway was activated in formononetin‑treated JME/CF15 cells, whereas treatment with the SIRT1 inhibitor, EX527, reversed the effects of formononetin on IL‑13‑induced inflammation and mucus formation in JME/CF15 cells. In conclusion, the findings of the current study indicated that formononetin may activate the SIRT1/Nrf2 signaling pathway, thereby inhibiting IL‑13‑induced inflammation and mucus formation in JME/CF15 cells. These results suggested that formononetin may represent a promising agent for the treatment of allergic rhinitis.
Collapse
Affiliation(s)
- Juanjuan Huang
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xianfeng Chen
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Aihua Xie
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
15
|
Wang WS, Zhao CS. Formononetin exhibits anticancer activity in gastric carcinoma cell and regulating miR-542-5p. Kaohsiung J Med Sci 2021; 37:215-225. [PMID: 33231363 DOI: 10.1002/kjm2.12322] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/12/2020] [Accepted: 10/18/2020] [Indexed: 12/14/2022] Open
Abstract
Formononetin exhibits anti-neoplastic activities in specific types of cancers, such as colon carcinoma and breast cancer. Nevertheless, its role in suppressing gastric carcinoma (GC) growth and metastatic-associated phenotypes has not been fully understood. Here, we demonstrated that formononetin decreased the viability of GC cell line SGC-7901 and MGC-803. Furthermore, formononetin suppressed the migration and invasion abilities of GC cells. Consistent with the results in vitro, the anticancer effect of formononetin was verified using xenograft model. The expression of microRNA-542-5p (miR-542-5p), acted as an oncogene in many cancers, was identified to be upregulated in GC. Importantly, miR-542-5p might involve in formononetin exhibits anticancer activity in GC cells. Taken together, these results indicate that formononetin inhibits the growth and aggressiveness of GC cells in vitro and in vivo.
Collapse
Affiliation(s)
- Wei-Song Wang
- Department of General Surgery, Zhuji Central Hospital, Zhuji, China
| | - Can-Song Zhao
- Department of General Surgery, Zhuji People's Hospital of Zhejiang, Zhuji, China
| |
Collapse
|
16
|
Zhang Y, Wang X. Targeting the Wnt/β-catenin signaling pathway in cancer. J Hematol Oncol 2020; 13:165. [PMID: 33276800 PMCID: PMC7716495 DOI: 10.1186/s13045-020-00990-3] [Citation(s) in RCA: 868] [Impact Index Per Article: 173.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
The aberrant Wnt/β-catenin signaling pathway facilitates cancer stem cell renewal, cell proliferation and differentiation, thus exerting crucial roles in tumorigenesis and therapy response. Accumulated investigations highlight the therapeutic potential of agents targeting Wnt/β-catenin signaling in cancer. Wnt ligand/ receptor interface, β-catenin destruction complex and TCF/β-catenin transcription complex are key components of the cascade and have been targeted with interventions in preclinical and clinical evaluations. This scoping review aims at outlining the latest progress on the current approaches and perspectives of Wnt/β-catenin signaling pathway targeted therapy in various cancer types. Better understanding of the updates on the inhibitors, antagonists and activators of Wnt/β-catenin pathway rationalizes innovative strategies for personalized cancer treatment. Further investigations are warranted to confirm precise and secure targeted agents and achieve optimal use with clinical benefits in malignant diseases.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.,Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China.,School of medicine, Shandong University, Jinan, 250021, Shandong, China.,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 250021, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,School of medicine, Shandong University, Jinan, 250021, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 250021, China.
| |
Collapse
|
17
|
Wang W, Ning J, He Y, Zhai L, Xiang F, Yao L, Ye L, Wu L, Ji T, Tang Z. Unveiling the mechanism of Astragalus membranaceus in the treatment of gastrointestinal cancers based on network pharmacology. Eur J Integr Med 2020. [DOI: 10.1016/j.eujim.2020.101249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
18
|
Nakonieczna S, Grabarska A, Kukula-Koch W. The Potential Anticancer Activity of Phytoconstituents against Gastric Cancer-A Review on In Vitro, In Vivo, and Clinical Studies. Int J Mol Sci 2020; 21:E8307. [PMID: 33167519 PMCID: PMC7663924 DOI: 10.3390/ijms21218307] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer belongs to the heterogeneous malignancies and, according to the World Health Organization, it is the fifth most commonly diagnosed cancer in men. The aim of this review is to provide an overview on the role of natural products of plant origin in the therapy of gastric cancer and to present the potentially active metabolites which can be used in the natural therapeutical strategies as the support to the conventional treatment. Many of the naturally spread secondary metabolites have been proved to exhibit chemopreventive properties when tested on the cell lines or in vivo. This manuscript aims to discuss the pharmacological significance of both the total extracts and the single isolated metabolites in the stomach cancer prevention and to focus on their mechanisms of action. A wide variety of plant-derived anticancer metabolites from different groups presented in the manuscript that include polyphenols, terpenes, alkaloids, or sulphur-containing compounds, underlines the multidirectional nature of natural products.
Collapse
Affiliation(s)
- Sylwia Nakonieczna
- Chair and Department of Pharmacognosy, Medical University of Lublin, 1, Chodzki str., 20-093 Lublin, Poland;
| | - Aneta Grabarska
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1, Chodźki, 20-093 Lublin, Poland
| | - Wirginia Kukula-Koch
- Chair and Department of Pharmacognosy, Medical University of Lublin, 1, Chodzki str., 20-093 Lublin, Poland;
| |
Collapse
|
19
|
Razavi R, Kenari RE, Farmani J, Jahanshahi M. Fabrication of zein/alginate delivery system for nanofood model based on pumpkin. Int J Biol Macromol 2020; 165:3123-3134. [PMID: 33127546 DOI: 10.1016/j.ijbiomac.2020.10.176] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/18/2020] [Accepted: 10/21/2020] [Indexed: 12/22/2022]
Abstract
Gastrectomy is among the most crucial types of surgeries proposed to treat gastric cancer and obesity. Gastrectomy patients experience difficulties such as energy deficit, anorexia, and malnutrition. The objective of the present study was to introduce nanofood as a fruitful strategy to supply the needed energy and nutrients for these patients and particularly control the release of proteins, lipids, and carbohydrates on the simulated gastrointestinal tract (GIT). Cooked pumpkin puree (CPP), sodium caseinate, sesame oil, rice bran oil, rice starch, sugar and pectin were applied to prepare oil in water nanoemulsion. Six delivery systems were prepared including various concentrations of zein (0.02-0.15% w/v) and alginate (0.01-0.16% w/v) in acidic (2.45-2.81) and alkaline (11.45-11.82) pH ranges. The particle size (83.5-207.0 nm) and calorific values (467.2-498.4 Cal/100 g) of samples were measured. Encapsulated food matrix nanoemulsion with zein/alginate's biopolymers delivery system (0.15:0.16 w/v, pH = 8.30) with 489.9 Cal/100 g exhibited the least digestible nutrients in the mouth (0.10%>) and gastric phase (6.91%>). It has high release nutrients in the small intestine phase (72.14%>). Therefore, it is introduced as the optimal formulation. The use of CPP in nanoemulsion formulation besides other ingredients is a good strategy to prepare nanofood for gastrectomy patients.
Collapse
Affiliation(s)
- Razie Razavi
- Department of Food Science and Technology, Sari Agricultural Sciences and Natural Resources University, Sari, Mazandaran 48181-68984, Iran
| | - Reza Esmaeilzadeh Kenari
- Department of Food Science and Technology, Sari Agricultural Sciences and Natural Resources University, Sari, Mazandaran 48181-68984, Iran.
| | - Jamshid Farmani
- Department of Food Science and Technology, Sari Agricultural Sciences and Natural Resources University, Sari, Mazandaran 48181-68984, Iran
| | - Mohsen Jahanshahi
- Department of Biotechnology, Babol Noshirvani University of Technology, Babol, Mazandaran 47148-71167, Iran
| |
Collapse
|
20
|
Song X, Fan J, Liu L, Liu X, Gao F. Coumarin derivatives with anticancer activities: An update. Arch Pharm (Weinheim) 2020; 353:e2000025. [DOI: 10.1002/ardp.202000025] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/15/2020] [Accepted: 04/22/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Xu‐Feng Song
- Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry and Chemical Engineering, College of Environmental and Energy EngineeringBeijing University of Technology Beijing China
| | - Jing Fan
- Hengshui University Hengshui Hebei China
| | - Lan Liu
- Medicine Vocational and Technical SchoolWuhan University Wuhan Hubei China
| | - Xiao‐Feng Liu
- Sinolite Industrial Co., Ltd. Hangzhou Zhejiang China
| | - Feng Gao
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP)Qilu University of Technology (Shandong Academy of Sciences) Jinan Shandong China
| |
Collapse
|
21
|
Formononetin inhibits tumor growth by suppression of EGFR-Akt-Mcl-1 axis in non-small cell lung cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:62. [PMID: 32276600 PMCID: PMC7146989 DOI: 10.1186/s13046-020-01566-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/26/2020] [Indexed: 12/22/2022]
Abstract
Background Epidermal growth factor receptor (EGFR) activating mutations play crucial roles in the tumorigenesis of human non-small cell lung cancer (NSCLC). The mechanism regarding how EGFR signaling regulates myeloid cell leukemia sequence 1 (Mcl-1) protein stability and ubiquitination remains undefined. Methods MTS assay was used for natural product library screening. The effect of formononetin (Formo) on NSCLC cells was determined by MTS assay and soft agar assay. Molecular modeling was performed to analyze the potential different binding modes between Formo and EGFR WT or mutants. Mcl-1 protein level and the inhibitory effect of Formo on EGFR signaling were examined by immunoblot, in vitro kinase assay, in vitro pulldown and ATP competition assays, co-immunoprecipitation assay, ubiquitination analysis, in vivo xenograft model, and immunohistochemical staining. Results Formo was identified as an EGFR inhibitor by a 98 commercially available natural product screening. Formo suppresses WT and mutant EGFR kinases activity in vitro, ex vivo, and in vivo. Molecular modeling indicates that Formo docks into the ATP-binding pocket of both WT and mutant EGFR. Formo inhibits EGFR-Akt signaling, which in turn activates GSK3β and promotes Mcl-1 phosphorylation in NSCLC cells. Treatment with Formo enhances the interaction between Mcl-1 and SCFFbw7, which eventually promotes Mcl-1 ubiquitination and degradation. Depletion of either GSK3β or SCFFbw7 compromised Formo-induced Mcl-1 downregulation. Finally, Formo inhibits the in vivo tumor growth in a xenograft mouse model. Conclusion This study highlights the importance of promoting ubiquitination-dependent Mcl-1 turnover might be an alternative strategy to enhance the anti-tumor efficacy of EGFR-TKI.
Collapse
|
22
|
Wang X, Zhao L, Ajay AK, Jiao B, Zhang X, Wang C, Gao X, Yuan Z, Liu H, Liu WJ. QiDiTangShen Granules Activate Renal Nutrient-Sensing Associated Autophagy in db/db Mice. Front Physiol 2019; 10:1224. [PMID: 31632286 PMCID: PMC6779835 DOI: 10.3389/fphys.2019.01224] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/09/2019] [Indexed: 12/24/2022] Open
Abstract
QiDiTangShen granules (QDTS) have been proven to reduce the proteinuria in patients with diabetic nephropathy (DN) effectively. The present study was aimed to investigate the mechanism underlying QDTS's renoprotection. The main components of QDTS were identified by ultra-high liquid chromatography-tandem mass spectrometry and pharmacological databases, among which active components were screened by oral bioavailability and drug-likeness. Their regulation on autophagy-related nutrient-sensing signal molecules (AMPK, SIRT1, and mTOR) was retrieved and analyzed through the Pubmed database. Then, db/db mice were randomly divided into three groups (model control, valsartan and QDTS), and given intragastric administration for 12 weeks, separately. Fasting and random blood glucose, body weight, urinary albumin excretion (UAE) and injury markers of liver and kidney were investigated to evaluate the effects and safety. Renal histological lesions were assessed, and the expressions of proteins related to nutrient-sensing signals and autophagy were investigated. Thirteen active components were screened from 78 components identified. Over half the components had already been reported to improve nutrient-sensing signals. QDTS significantly reduced UAE, ameliorated mesangial matrix deposition, alleviate the expression of protein and mRNA of TGF-β, α-SMA, and Col I, as well as improved the quality of mitochondria and the number of autophagic vesicles of renal tubular cells although the blood glucose was not decreased in db/db mice. Compared to the db/db group, the expression of the autophagy-inducible protein (Atg14 and Beclin1) and microtubule-associated protein 1 light chain 3-II (LC3-II) were up-regulated, autophagic substrate transporter p62 was down-regulated in QDTS group. It was also found that the expression of SIRT1 and the proportion of p-AMPK (thr172)/AMPK were increased, while the p-mTOR (ser2448)/mTOR ratio was decreased after QDTS treatment in db/db mice, which was consistent with the effect of its active ingredients on the nutrient-sensing signal pathway as reported previously. Therefore, QDTS may prevent the progression of DN by offering the anti-fibrotic effect. The renoprotection is probably attributable to the regulation of nutrient-sensing signal pathways, which activates autophagy.
Collapse
Affiliation(s)
- Xiangming Wang
- Department of Endocrinology and Nephrology, Renal Research Institute of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Li Zhao
- Department of Endocrinology and Nephrology, Renal Research Institute of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Amrendra K. Ajay
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Baihai Jiao
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Xianhui Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Health Management Center, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Gao
- Department of Endocrinology and Nephrology, Renal Research Institute of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhongyu Yuan
- Department of Endocrinology and Nephrology, Renal Research Institute of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Hongfang Liu
- Department of Endocrinology and Nephrology, Renal Research Institute of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Department of Endocrinology and Nephrology, Renal Research Institute of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|