1
|
Verma R, Dash S, Ankita, Thakur S, Kumar R, Singh G, Kaur C. Genus Bauhinia (Fabaceae): A review from phytochemistry to pharmacology- Exploring traditional uses and toxicological insights across Asia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156246. [PMID: 39571414 DOI: 10.1016/j.phymed.2024.156246] [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: 03/27/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND The genus Bauhinia and its species has been known since ages by tribal people and medicinal practitioners in tropical and subtropical regions for the treatment of diabetes, diarrhea, cough, fever, stomach disorders, skin diseases, and other diseases. STUDY DESIGN Our aim was to collect and explore the literature available on the traditional applications and medicinal potential of genus Bauhinia across Asia, so that this review can be used as standard to analyze the immense potential of this genus and can be explored further for clinical use. METHODS The information was rigorously gathered from Google Scholar, Pub Med, Elsevier, Wiley Online Search, Science Direct, and other literature sources. RESULTS As per the literature, this genus possesses antimicrobial, antioxidant, nephroprotective, anticancer, hepatoprotective, antidiabetic, anti-inflammatory, and antidepressant activities both in vitro and in vivo, due to the presence of flavonoids, steroidal saponins, bauhinioxepins, chromanones, and phenolic compounds. CONCLUSION In this review, we have detailed for the first time the categorized information about traditional uses, geographical distribution, morphological features, phytochemistry, pharmacological, and toxicological effects, and patents associated with Bauhinia species. However, more research is needed to explore the mechanisms of action, pharmacokinetics of the phytoconstituents, and clinical evaluation for their future use in treating various ailments.
Collapse
Affiliation(s)
- Rupali Verma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab
| | - Shubham Dash
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab
| | - Ankita
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab
| | - Shorya Thakur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab
| | - Gurvinder Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab.
| |
Collapse
|
2
|
Demir K, Turgut R, Şentürk S, Işıklar H, Günalan E. The Therapeutic Effects of Bioactive Compounds on Colorectal Cancer via PI3K/Akt/mTOR Signaling Pathway: A Critical Review. Food Sci Nutr 2024; 12:9951-9973. [PMID: 39723045 PMCID: PMC11666977 DOI: 10.1002/fsn3.4534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 12/28/2024] Open
Abstract
Understanding the molecular signaling pathways of colorectal cancer (CRC) can be accepted as the first step in treatment strategy. Permanent mTOR signaling activation stimulates the CRC process via various biological processes. It supplies the survival of CRC stem cells, tumorigenesis, morbidity, and decreased response to drugs in CRC pathogenesis. Therefore, inhibition of the mTOR signaling by numerous bioactive components may be effective against CRC. The study aims to discuss the therapeutic capacity of various polyphenols, terpenoids, and alkaloids on CRC via the PI3K/Akt/mTOR pathway. The potential molecular effects of bioactive compounds on the mTOR pathway's upstream and downstream targets are examined. Each bioactive component causes various physiological processes, such as triggering free radical production, disruption of mitochondrial membrane potential, cell cycle arrest, inhibition of CRC stem cell migration, and suppression of glycolysis through mTOR signaling inhibition. As a result, carcinogenesis is inhibited by inducing apoptosis and autophagy. However, it should be noted that studies are primarily in vitro dose-dependent treatment researchers. This study raises awareness about the role of phenolic compounds in treating CRC, contributing to their future use as anticancer agents. These bioactive compounds have the potential to be developed into food supplementation to prevent and treat various cancer types including CRC. This review has the potential to lead to further development of clinical studies. In the future, mTOR inhibition by applying several bioactive agents using advanced drug delivery systems may contribute to CRC treatment with 3D cell culture and in vivo clinical studies.
Collapse
Affiliation(s)
- Kübra Demir
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
- Faculty of Health Science, Department of Nutrition and DieteticsSabahattin Zaim UniversityIstanbulTürkiye
| | - Rana Turgut
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
| | - Selcen Şentürk
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
| | - Handan Işıklar
- Faculty of Medicine, Department of Internal MedicineYalova UniversityYalovaTürkiye
| | - Elif Günalan
- Faculty of Health Science, Department of Nutrition and DieteticsIstanbul Health and Technology UniversityIstanbulTürkiye
| |
Collapse
|
3
|
Martiniakova M, Penzes N, Biro R, Sarocka A, Kovacova V, Mondockova V, Ciernikova S, Omelka R. Sea buckthorn and its flavonoids isorhamnetin, quercetin, and kaempferol favorably influence bone and breast tissue health. Front Pharmacol 2024; 15:1462823. [PMID: 39444603 PMCID: PMC11497132 DOI: 10.3389/fphar.2024.1462823] [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: 07/10/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Bone tissue and breast tissue are interrelated, as demonstrated by breast microcalcifications, breast cancer bone metastases, bone morphogenetic proteins, and Wnt signaling. In addition, osteoblasts and osteoclasts represent an important switch of tumor cell dormancy during bone metastasis. Damage to both types of tissues mentioned above is highly prevalent, especially in postmenopausal women, and manifests itself in osteoporosis and breast cancer. Sea buckthorn (Elaeagnus rhamnoides L.), a botanical drug with high antioxidant, antitumor, anti-inflammatory, immunomodulatory, and regenerative properties, has great therapeutic potential due to the unique composition of its bioactive metabolites. This review aimed to summarize the current knowledge from in vitro and in vivo studies on the effect of sea buckthorn, as well as its most widespread flavonoids isorhamnetin, quercetin, and kaempferol, on bone and breast tissue health. In vitro studies have revealed the beneficial impacts of sea buckthorn and aforementioned flavonoids on both bone health (bone remodeling, mineralization, and oxidative stress) and breast tissue health (cancer cell proliferation, apoptosis, tumor growth, and metastatic behavior). In vivo studies have documented their protective effects against disturbed bone microarchitecture and reduced bone strength in animal models of osteoporosis, as well as against tumor expansion and metastatic properties in animal xenograft models. In any case, further research and clinical trials are needed to carefully evaluate the potential therapeutic benefits of sea buckthorn and its flavonoids. Based on the available information, however, it can be concluded that these bioactive metabolites favorably affect both bone and breast tissue health.
Collapse
Affiliation(s)
- Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Noemi Penzes
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Roman Biro
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Anna Sarocka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| |
Collapse
|
4
|
Xu D, Yuan L, Meng F, Lu D, Che M, Yang Y, Liu W, Nan Y. Research progress on antitumor effects of sea buckthorn, a traditional Chinese medicine homologous to food and medicine. Front Nutr 2024; 11:1430768. [PMID: 39045282 PMCID: PMC11263281 DOI: 10.3389/fnut.2024.1430768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024] Open
Abstract
Sea buckthorn (Hippophae Fructus), as a homologous species of medicine and food, is widely used by Mongolians and Tibetans for its anti-tumor, antioxidant and liver-protecting properties. In this review, the excellent anti-tumor effect of sea buckthorn was first found through network pharmacology, and its active components such as isorhamnetin, quercetin, gallic acid and protocatechuic acid were found to have significant anti-tumor effects. The research progress and application prospect of sea buckthorn and its active components in anti-tumor types, mechanism of action, liver protection, anti-radiation and toxicology were reviewed, providing theoretical basis for the development of sea buckthorn products in the field of anti-tumor research and clinical application.
Collapse
Affiliation(s)
- Duojie Xu
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Fandi Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Doudou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Mengying Che
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yating Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Wenjing Liu
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yi Nan
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
5
|
Biswas P, Kaium MA, Islam Tareq MM, Tauhida SJ, Hossain MR, Siam LS, Parvez A, Bibi S, Hasan MH, Rahman MM, Hosen D, Islam Siddiquee MA, Ahmed N, Sohel M, Azad SA, Alhadrami AH, Kamel M, Alamoudi MK, Hasan MN, Abdel-Daim MM. The experimental significance of isorhamnetin as an effective therapeutic option for cancer: A comprehensive analysis. Biomed Pharmacother 2024; 176:116860. [PMID: 38861855 DOI: 10.1016/j.biopha.2024.116860] [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: 04/06/2024] [Revised: 05/26/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
Isorhamnetin (C16H12O7), a 3'-O-methylated derivative of quercetin from the class of flavonoids, is predominantly present in the leaves and fruits of several plants, many of which have traditionally been employed as remedies due to its diverse therapeutic activities. The objective of this in-depth analysis is to concentrate on Isorhamnetin by addressing its molecular insights as an effective anticancer compound and its synergistic activity with other anticancer drugs. The main contributors to Isorhamnetin's anti-malignant activities at the molecular level have been identified as alterations of a variety of signal transduction processes and transcriptional agents. These include ROS-mediated cell cycle arrest and apoptosis, inhibition of mTOR and P13K pathway, suppression of MEK1, PI3K, NF-κB, and Akt/ERK pathways, and inhibition of Hypoxia Inducible Factor (HIF)-1α expression. A significant number of in vitro and in vivo research studies have confirmed that it destroys cancerous cells by arresting cell cycle at the G2/M phase and S-phase, down-regulating COX-2 protein expression, PI3K, Akt, mTOR, MEK1, ERKs, and PI3K signaling pathways, and up-regulating apoptosis-induced genes (Casp3, Casp9, and Apaf1), Bax, Caspase-3, P53 gene expression and mitochondrial-dependent apoptosis pathway. Its ability to suppress malignant cells, evidence of synergistic effects, and design of drugs based on nanomedicine are also well supported to treat cancer patients effectively. Together, our findings establish a crucial foundation for understanding Isorhamnetin's underlying anti-cancer mechanism in cancer cells and reinforce the case for the requirement to assess more exact molecular signaling pathways relating to specific cancer and in vivo anti-cancer activities.
Collapse
Affiliation(s)
- Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh; ABEx Bio-Research Center, East Azampur, Dhaka 1230, Bangladesh
| | - Md Abu Kaium
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Mohaimenul Islam Tareq
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Sadia Jannat Tauhida
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Ridoy Hossain
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Labib Shahriar Siam
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Anwar Parvez
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1216, Bangladesh
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad 41000, Pakistan
| | - Md Hasibul Hasan
- Department of Food Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj 8100, Bangladesh
| | - Md Moshiur Rahman
- Department of Information Systems Security, Faculty of Science & Technology, Bangladesh University of Professionals, Mirpur 1216, Bangladesh
| | - Delwar Hosen
- Department of Electrical and Computer Engineering, North South University, Dhaka 1229, Bangladesh
| | | | - Nasim Ahmed
- Department of Pharmacy, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Sohel
- Department of Biochemistry and Molecular Biology, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Salauddin Al Azad
- Immunoinformatics and Vaccinomics Research Unit, RPG Interface Lab, Jashore 7400, Bangladesh
| | - Albaraa H Alhadrami
- Faculty of Medicine, King Abdulaziz University, P.O.Box 80402, Jeddah 21589, Saudi Arabia
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Mariam K Alamoudi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Md Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh.
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
6
|
Ayachi A, Boy G, Samet S, Téné N, Bouzayani B, Treilhou M, Mezghani-Jarraya R, Billet A. Isolation, NMR Characterization, and Bioactivity of a Flavonoid Triglycoside from Anthyllis henoniana Stems: Antioxidant and Antiproliferative Effects on MDA-MB-231 Breast Cancer Cells. Antioxidants (Basel) 2024; 13:793. [PMID: 39061863 PMCID: PMC11273540 DOI: 10.3390/antiox13070793] [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: 06/06/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Plant extracts are considered as a large source of active biomolecules, especially in phytosanitary and pharmacological fields. Anthyllis henoniana is a woody Saharan plant located in the big desert of North Africa. Our previous research paper proved the richness of the methanol extract obtained from the stems in flavonoids and phenolic compounds as well as its remarkable antioxidant activity. In this research, we started by investigating the phytochemical composition of the methanol extract using high performance liquid chromatography coupled with electrospray ionization mass spectrometry (LC-MS/MS). Among the 41 compounds identified, we isolated and characterized (structurally and functionally) the most abundant product, a flavonoid triglycoside (AA770) not previously described in this species. This compound, which presents no cytotoxic activity, exhibits an interesting cellular antioxidant effect by reducing reactive oxygen species (ROS) generation, and an antiproliferative action on breast cancer cells. This study provides a preliminary investigation into the pharmacological potential of the natural compound AA770, isolated and identified from Anthyllis henoniana for the first time.
Collapse
Affiliation(s)
- Amani Ayachi
- Laboratory of Organic Chemistry LR17ES08, Natural Substances Team, Faculty of Sciences of Sfax, University of Sfax, P.O. Box 1171, Sfax 3000, Tunisia; (A.A.); (S.S.); (B.B.)
| | - Guillaume Boy
- Equipe BTSB-EA 7417, Institut National Universitaire Jean-François Champollion, Université de Toulouse, Place de Verdun, 81012 Albi, France; (G.B.); (N.T.); (M.T.)
| | - Sonda Samet
- Laboratory of Organic Chemistry LR17ES08, Natural Substances Team, Faculty of Sciences of Sfax, University of Sfax, P.O. Box 1171, Sfax 3000, Tunisia; (A.A.); (S.S.); (B.B.)
| | - Nathan Téné
- Equipe BTSB-EA 7417, Institut National Universitaire Jean-François Champollion, Université de Toulouse, Place de Verdun, 81012 Albi, France; (G.B.); (N.T.); (M.T.)
| | - Bouthaina Bouzayani
- Laboratory of Organic Chemistry LR17ES08, Natural Substances Team, Faculty of Sciences of Sfax, University of Sfax, P.O. Box 1171, Sfax 3000, Tunisia; (A.A.); (S.S.); (B.B.)
| | - Michel Treilhou
- Equipe BTSB-EA 7417, Institut National Universitaire Jean-François Champollion, Université de Toulouse, Place de Verdun, 81012 Albi, France; (G.B.); (N.T.); (M.T.)
| | - Raoudha Mezghani-Jarraya
- Laboratory of Organic Chemistry LR17ES08, Natural Substances Team, Faculty of Sciences of Sfax, University of Sfax, P.O. Box 1171, Sfax 3000, Tunisia; (A.A.); (S.S.); (B.B.)
| | - Arnaud Billet
- Equipe BTSB-EA 7417, Institut National Universitaire Jean-François Champollion, Université de Toulouse, Place de Verdun, 81012 Albi, France; (G.B.); (N.T.); (M.T.)
| |
Collapse
|
7
|
Lei J, Yang J, Bao C, Lu F, Wu Q, Wu Z, Lv H, Zhou Y, Liu Y, Zhu N, Yu Y, Zhang Z, Hu M, Lin L. Isorhamnetin: what is the in vitro evidence for its antitumor potential and beyond? Front Pharmacol 2024; 15:1309178. [PMID: 38650631 PMCID: PMC11033395 DOI: 10.3389/fphar.2024.1309178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
Isorhamnetin (ISO) is a phenolic compound belonging to flavonoid family, showcasing important in vitro pharmacological activities such as antitumor, anti-inflammation, and organ protection. ISO is predominantly extracted from Hippophae rhamnoides L. This plant is well-known in China and abroad because of its "medicinal and food homologous" characteristics. As a noteworthy natural drug candidate, ISO has received considerable attention in recent years owing to its low cost, wide availability, high efficacy, low toxicity, and minimal side effects. To comprehensively elucidate the multiple biological functions of ISO, particularly its antitumor activities and other pharmacological potentials, a literature search was conducted using electronic databases including Web of Science, PubMed, Google Scholar, and Scopus. This review primarily focuses on ISO's ethnopharmacology. By synthesizing the advancements made in existing research, it is found that the general effects of ISO involve a series of in vitro potentials, such as antitumor, protection of cardiovascular and cerebrovascular, anti-inflammation, antioxidant, and more. This review illustrates ISO's antitumor and other pharmacological potentials, providing a theoretical basis for further research and new drug development of ISO.
Collapse
Affiliation(s)
- Jiaming Lei
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Jianbao Yang
- School of Public Health, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Cuiyu Bao
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular and Metabolic Disorder, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Feifei Lu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qing Wu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zihan Wu
- School of Biomedical Engineering, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Hong Lv
- School of Public Health, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanhong Zhou
- Department of Medical School of Facial Features, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yifei Liu
- School of Biomedical Engineering, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ni Zhu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - You Yu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhipeng Zhang
- Department of Medical School of Facial Features, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Li Lin
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
8
|
Lin X, Chi W, Geng X, Jiang Q, Ma B, Dai B, Sui Y, Jiang J. Evaluation of the Mechanism of Yishan Formula in Treating Breast Cancer Based on Network Pharmacology and Experimental Verification. Comb Chem High Throughput Screen 2024; 27:2583-2597. [PMID: 38178684 DOI: 10.2174/0113862073266004231105164321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/07/2023] [Accepted: 08/31/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Yishan formula (YSF) has a significant effect on the treatment of breast cancer, which can improve the quality of life and prolong the survival of patients with breast cancer; however, its mechanism of action is unknown. OBJECTIVE In this study, network pharmacology and molecular docking methods have been used to explore the potential pharmacological effects of the YSF, and the predicted targets have been validated by in vitro experiments. METHODS Active components and targets of the YSF were obtained from the TCMSP and Swiss target prediction website. Four databases, namely GeneCards, OMIM, TTD, and DisGeNET, were used to search for disease targets. The Cytoscape v3.9.0 software was utilized to draw the network of drug-component-target and selected core targets. DAVID database was used to analyze the biological functions and pathways of key targets. Finally, molecular docking and in vitro experiments have been used to verify the hub genes. RESULTS Through data collection from the database, 157 active components and 618 genes implicated in breast cancer were obtained and treated using the YSF. After screening, the main active components (kaempferol, quercetin, isorhamnetin, dinatin, luteolin, and tamarixetin) and key genes (AKT1, TP53, TNF, IL6, EGFR, SRC, VEGFA, STAT3, MAPK3, and JUN) were obtained. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated that the YSF could affect the progression of breast cancer by regulating biological processes, such as signal transduction, cell proliferation and apoptosis, protein phosphorylation, as well as PI3K-Akt, Rap1, MAPK, FOXO, HIF-1, and other related signaling pathways. Molecular docking suggested that IL6 with isorhamnetin, MAPK3 with kaempferol, and EGFR with luteolin have strong binding energy. The experiment further verified that YSF can control the development of breast cancer by inhibiting the expression of the hub genes. CONCLUSION This study showed that resistance to breast cancer may be achieved by the synergy of multiple active components, target genes, and signal pathways, which can provide new avenues for breast cancer-targeted therapy.
Collapse
Affiliation(s)
- Xiaoyue Lin
- Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Wencheng Chi
- Heilongjiang University of Chinese Medicine, Harbin, 150000, China
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150000, China
| | - Xue Geng
- Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Qinghui Jiang
- Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Baozhu Ma
- Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Bowen Dai
- Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Yutong Sui
- Shenzhen Hospital of Southern Medical University, Shenzhen, 518110, China
| | - Jiakang Jiang
- Heilongjiang University of Chinese Medicine, Harbin, 150000, China
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150000, China
| |
Collapse
|
9
|
Zhang P, Fan L, Zhang D, Zhang Z, Wang W. In Vitro Anti-Tumor and Hypoglycemic Effects of Total Flavonoids from Willow Buds. Molecules 2023; 28:7557. [PMID: 38005279 PMCID: PMC10673267 DOI: 10.3390/molecules28227557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Salix babylonica L. is a species of willow tree that is widely cultivated worldwide as an ornamental plant, but its medicinal resources have not yet been reasonably developed or utilized. Herein, we extracted and purified the total flavonoids from willow buds (PTFW) for component analysis in order to evaluate their in vitro anti-tumor and hypoglycemic activities. Through Q-Orbitrap LC-MS/MS analysis, a total of 10 flavonoid compounds were identified (including flavones, flavan-3-ols, and flavonols). The inhibitory effects of PTFW on the proliferation of cervical cancer HeLa cells, colon cancer HT-29 cells, and breast cancer MCF7 cells were evaluated using an MTT assay. Moreover, the hypoglycemic activity of PTFW was determined by investigating the inhibitory effects of PTFW on α-amylase and α-glucosidase. The results indicated that PTFW significantly suppressed the proliferation of HeLa cells, HT-29 cells, and MCF7 cells, with IC50 values of 1.432, 0.3476, and 2.297 mg/mL, respectively. PTFW, at different concentrations, had certain inhibitory effects on α-amylase and α-glucosidase, with IC50 values of 2.94 mg/mL and 1.87 mg/mL, respectively. In conclusion, PTFW at different doses exhibits anti-proliferation effects on all three types of cancer cells, particularly on HT-29 cells, and also shows significant hypoglycemic effects. Willow buds have the potential to be used in functional food and pharmaceutical industries.
Collapse
Affiliation(s)
- Peng Zhang
- College of Life Engineering, Shenyang Institute of Technology, Fushun 113122, China; (L.F.); (D.Z.)
| | - Lulu Fan
- College of Life Engineering, Shenyang Institute of Technology, Fushun 113122, China; (L.F.); (D.Z.)
| | - Dongyan Zhang
- College of Life Engineering, Shenyang Institute of Technology, Fushun 113122, China; (L.F.); (D.Z.)
| | - Zehui Zhang
- College of Laboratory Animal Medicine and Science, Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China;
| | - Weili Wang
- Liao Ning Institute for Drug Control, Shenyang 110031, China
| |
Collapse
|
10
|
Wendlocha D, Krzykawski K, Mielczarek-Palacz A, Kubina R. Selected Flavonols in Breast and Gynecological Cancer: A Systematic Review. Nutrients 2023; 15:2938. [PMID: 37447264 DOI: 10.3390/nu15132938] [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: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
The consumption of foods that are rich in phenolic compounds has chemopreventive effects on many cancers, including breast cancer, ovarian cancer, and endometrial cancer. A wide spectrum of their health-promoting properties such as antioxidant, anti-inflammatory, and anticancer activities, has been demonstrated. This paper analyzes the mechanisms of the anticancer action of selected common flavonols, including kemferol, myricetin, quercetin, fisetin, galangin, isorhamnetin, and morin, in preclinical studies, with particular emphasis on in vitro studies in gynecological cancers and breast cancer. In the future, these compounds may find applications in the prevention and treatment of gynecological cancers and breast cancer, but this requires further, more advanced research.
Collapse
Affiliation(s)
- Dominika Wendlocha
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Kamil Krzykawski
- Silesia LabMed: Centre for Research and Implementation, Medical University of Silesia in Katowice, 40-752 Katowice, Poland
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Robert Kubina
- Silesia LabMed: Centre for Research and Implementation, Medical University of Silesia in Katowice, 40-752 Katowice, Poland
- Department of Pathology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| |
Collapse
|
11
|
Tiwari A, Tiwari V, Sharma A, Singh D, Singh Rawat M, Virmani T, Virmani R, Kumar G, Kumar M, Alhalmi A, Noman OM, Mothana RA, Alali M. Tanshinone-I for the treatment of uterine fibroids: Molecular docking, simulation, and density functional theory investigations. Saudi Pharm J 2023; 31:1061-1076. [PMID: 37250358 PMCID: PMC10209546 DOI: 10.1016/j.jsps.2023.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/01/2023] [Indexed: 05/31/2023] Open
Abstract
Uterine fibroids (UF), most prevalent gynecological disorder, require surgery when symptomatic. It is estimated that between 25 and 35 percent of women wait until the symptoms have worsened like extended heavy menstrual bleeding and severe pelvic pain. These UF may be reduced in size through various methods such as medical or surgical intervention. Progesterone (prog) is a crucial hormone that restores the endometrium and controls uterine function. In the current study, 28 plant-based molecules are identified from previous literature and docked onto the prog receptors with 1E3K and 2OVH. Tanshinone-I has shown the best docking score against both proteins. The synthetic prog inhibitor Norethindrone Acetate is used as a standard to evaluate the docking outcomes. The best compound, tanshinone-I, was analyzed using molecular modeling and DFT. The RMSD for the 1E3K protein-ligand complex ranged from 0.10 to 0.42 Å, with an average of 0.21 Å and a standard deviation (SD) of 0.06, while the RMSD for the 2OVH protein-ligand complex ranged from 0.08 to 0.42 Å, with an average of 0.20 Å and a SD of 0.06 showing stable interaction. In principal component analysis, the observed eigen values of HPR-Tanshinone-I fluctuate between -1.11 to 1.48 and -1.07 to 1.25 for PC1 and PC2, respectively (1E3K), and the prog-tanshinone-I complex shows eigen values of -38.88 to -31.32 and -31.32 to 35.87 for PC1 and PC2, respectively (2OVH), which shows Tanshinone-I forms a stable protein-ligand complex with 1E3K in comparison to 2OVH. The Free Energy Landscape (FEL) analysis shows the Gibbs free energy in the range of 0 to 8 kJ/mol for Tanshinone-I with 1E3K and 0 to 14 kJ/mol for Tanshinone-I with the 2OVH complex. The DFT calculation reveals ΔE value of 2.8070 eV shows tanshinone-I as a stable compound. 1E3K modulates the prog pathway, it may have either an agonistic or antagonistic effect on hPRs. Tanshinone-I can cause ROS, apoptosis, autophagy (p62 accumulation), up-regulation of inositol requiring protein-1, enhancer-binding protein homologous protein, p-c-Jun N-terminal kinase (p-JNK), and suppression of MMPs. Bcl-2 expression can change LC3I to LC3II and cause apoptosis through Beclin-1 expression.
Collapse
Affiliation(s)
- Abhishek Tiwari
- Department of Pharmacy, Pharmacy Academy, IFTM University, Lodhipur-Rajpur, Moradabad 244102, India
| | - Varsha Tiwari
- Department of Pharmacy, Pharmacy Academy, IFTM University, Lodhipur-Rajpur, Moradabad 244102, India
| | - Ajay Sharma
- Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravi Shankar Shukla University, Raipur, Chhattisgarh, India
| | - Manju Singh Rawat
- University Institute of Pharmacy, Pt. Ravi Shankar Shukla University, Raipur, Chhattisgarh, India
| | - Tarun Virmani
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India
| | - Reshu Virmani
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India
| | - Girish Kumar
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India
| | - Manish Kumar
- School of Pharmaceutical Sciences, CT University, Ludhiana- 142024 Punjab, India
| | - Abdulsalam Alhalmi
- Department of Pharmaceutical Sciences, College of Pharmacy, Aden University, Aden, Yemen
| | - Omar M. Noman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ramzi A. Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Alali
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany
| |
Collapse
|
12
|
Yang T, Xiao Y, Liu S, Luo F, Tang D, Yu Y, Xie Y. Isorhamnetin induces cell cycle arrest and apoptosis by triggering DNA damage and regulating the AMPK/mTOR/p70S6K signaling pathway in doxorubicin-resistant breast cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154780. [PMID: 37004402 DOI: 10.1016/j.phymed.2023.154780] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/16/2023] [Accepted: 03/19/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Acquired resistance to doxorubicin (DOX) inevitably limits its clinical use against breast cancer (BC). Isorhamnetin (IS), a native flavonoid which extensively available in vegetables, fruits, and phytomedicine, has been deemed to the probable cancer chemopreventive agent in preceding explorations since it exhibits satisfied antitumor activity. So far, the strategy for alleviating DOX resistance by using IS as a sensitizer against resistant BC has not yet been covered. PURPOSE To investigate the effect of IS on potentiating the chemoreceptivity of drug-resistant BC cells to DOX in vitro and in vivo and elucidate the possible molecular mechanisms. METHODS MTS assays, colony formation assays, three-dimensional (3D) tumor spheroid model, and migration assay were deployed to verify the inhibiting action of IS in the presence or absence of DOX on resistant BC cells in vitro. Apoptosis, cell cycle regulation, and endocellular reactive oxygen species (ROS) were determined by flow cytometry. Protein levels were monitored by western blotting. Nuclear staining and EdU proliferation were photographed with a confocal laser scanning microscope. The effects of the IS and DOX combination on the tumorigenesis in the xenograft experiments were evaluated for further confirming the in vitro cytotoxicity. RESULTS IS significantly inhibited cell proliferation and migration and enhanced the antitumor competence of DOX against resistant BC cells both in vitro and in vivo. Adjuvant IS (50 μM) effectively enhanced the proapoptotic impacts of DOX in resistant BC cells (35.38 ± 3.18%, vs. 5.83 ± 0.68% in the DOX group) by suppressing the expression of bcl 2 in addition to enhancing cleaved caspase 3, ultimately leading to DNA condensation and fragmentation. IS (20, 30, and 50 μM) treatments induced significant increases in the G2/M populations (41.60 ± 1.28%, 44.60 ± 1.14%, and 50.64 ± 0.67%, vs. 35.84 ± 1.56% in the untreated control in MCF7/ADR cells, p < 0.01) via regulating CDK1/Cyclin B1 complex expression, subsequently triggering the inhibition of BC proliferation. In addition, IS (10, 20, 30, and 50 μM) stimulated the production of interstitial ROS in MCF7/ADR cells, by 3.99-, 4.20-, 6.29-, and 6.78-fold, respectively, versus the untreated group (p < 0.001), which were involved in DNA damage and AMPK-caused intercept of the mTOR/p70S6K signaling. CONCLUSION Our study suggested the anti-breast cancer actions of IS as a DOX sensitizer and expounded the underlying molecular mechanisms, showing that IS could be deemed to a capable alternative for resistant BC cure.
Collapse
Affiliation(s)
- Tianshu Yang
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Yi Xiao
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Shuo Liu
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Fazhen Luo
- Pharmacy Department, Shanghai Integrated traditional Chinese and Western Medicine Hospital, Shanghai 200082, China
| | - Dongyun Tang
- Pharmacy Department, Xiangshan Hospital of Traditional Chinese Medicine, Shanghai 200020, China
| | - Yilin Yu
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Yan Xie
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
13
|
Sarkar S, Das AK, Bhattacharya S, Gachhui R, Sil PC. Isorhamnetin exerts anti-tumor activity in DEN + CCl 4-induced HCC mice. Med Oncol 2023; 40:188. [PMID: 37226027 DOI: 10.1007/s12032-023-02050-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/06/2023] [Indexed: 05/26/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most prevalent type of liver cancer and the main cause of cancer death globally. The use of medicinal herbs as chemotherapeutic agents in cancer treatment is receiving attention as they possess no or minimum side effects. Isorhamnetin (IRN), a flavonoid, has been under attention for its anti-inflammatory and anti-proliferative properties in a number of cancers, including colorectal, skin, and lung cancers. However, the in vivo mechanism of isorhamnetin to suppress liver cancer has yet to be explored. METHODS AND RESULT HCC was induced by N-diethylnitrosamine (DEN) and carbon tetrachloride (CCL4) in Swiss albino mice. Isorhamnetin (100 mg/kg body weight) was given to examine its anti-tumor properties in HCC mice model. Histological analysis and liver function assays were performed to assess changes in liver anatomy. Probable molecular pathways were explored using immunoblot, qPCR, ELISA, and immunohistochemistry techniques. Isorhamnetin inhibited various pro-inflammatory cytokines to suppress cancer-inducing inflammation. Additionally, it regulated Akt and MAPKs to suppress Nrf2 signaling. Isorhamnetin activated PPAR-γ and autophagy while suppressing cell cycle progression in DEN + CCl4-administered mice. Additionally, isorhamnetin regulated various signaling pathways to suppress cell proliferation, metabolism, and epithelial-mesenchymal transition in HCC. CONCLUSION Regulating diverse cellular signaling pathways makes isorhamnetin a better anti-cancer chemotherapeutic candidate in HCC. Importantly, the anti-TNF-α properties of isorhamnetin could prove it a valuable therapeutic agent in sorafenib-resistant HCC patients. Additionally, anti-TGF-β properties of isorhamnetin could be utilized to reduce the EMT-inducing side effects of doxorubicin.
Collapse
Affiliation(s)
- Sayanta Sarkar
- Department of Life Sciences & Biotechnology, Jadavpur University, 188, Raja SC Mullick Road, Kolkata, 700032, India
| | - Abhishek Kumar Das
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal, 700054, India
| | - Semantee Bhattacharya
- Indian Association for the Cultivation of Science, 2A & 2B, Raja Subodh Chandra Mallick Rd, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Ratan Gachhui
- Department of Life Sciences & Biotechnology, Jadavpur University, 188, Raja SC Mullick Road, Kolkata, 700032, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
14
|
Lv Y, Mou Y, Su J, Liu S, Ding X, Yuan Y, Li G, Li G. The inhibitory effect and mechanism of Resina Draconis on the proliferation of MCF-7 breast cancer cells: a network pharmacology-based analysis. Sci Rep 2023; 13:3816. [PMID: 36882618 PMCID: PMC9992681 DOI: 10.1038/s41598-023-30585-0] [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: 11/09/2022] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Resina Draconis (RD) is known as the "holy medicine for promoting blood circulation" and possesses antitumor properties against various types of cancer, including breast cancer (BC); however, the underlying mechanism is not well understood. To explore the potential mechanism of RD against BC using network pharmacology and experimental validation, data on bioactive compounds, potential targets of RD, and related genes of BC were obtained from multiple public databases. Gene Ontology (GO) and KEGG pathway analyses were performed via the DAVID database. Protein interactions were downloaded from the STRING database. The mRNA and protein expression levels and survival analysis of the hub targets were analyzed using the UALCAN, HPA, Kaplan‒Meier mapper, and cBioPortal databases. Subsequently, molecular docking was used to verify the selected key ingredients and hub targets. Finally, the predicted results of network pharmacology methods were verified by cell experiments. In total, 160 active ingredients were obtained, and 148 RD target genes for the treatment of BC were identified. KEGG pathway analysis indicated that RD exerted its therapeutic effects on BC by regulating multiple pathways. Of these, the PI3K-AKT pathway was indicated to play an important role. In addition, RD treatment of BC seemed to involve the regulation of hub targets that were identified based on PPI interaction network analysis. Validation in different databases showed that AKT1, ESR1, HSP90AA1, CASP3, SRC and MDM2 may be involved in the carcinogenesis and progression of BC and that ESR1, IGF1 and HSP90AA1 were correlated with worse overall survival (OS) in BC patients. Molecular docking results showed that 103 active compounds have good binding activity with the hub targets, among which flavonoid compounds were the most important active components. Therefore, the sanguis draconis flavones (SDF) were selected for subsequent cell experiments. The experimental results showed that SDF significantly inhibited the cell cycle and cell proliferation of MCF-7 cells through the PI3K/AKT pathway and induced MCF-7 cell apoptosis. This study has preliminarily reported on the active ingredients, potential targets, and molecular mechanism of RD against BC, and RD was shown to exert its therapeutic effects on BC by regulating the PI3K/AKT pathway and related gene targets. Importantly, our work could provide a theoretical basis for further study of the complex anti-BC mechanism of RD.
Collapse
Affiliation(s)
- Yana Lv
- Yunnan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong, 666100, China.,Yunnan Key Laboratory of Southern Medicinal Utilization, Jinghong, 666100, China
| | - Yan Mou
- Yuxi Normal University, Yuxi, 653100, China
| | - Jing Su
- Yunnan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong, 666100, China.,Yunnan Key Laboratory of Southern Medicinal Utilization, Jinghong, 666100, China
| | - Shifang Liu
- Yunnan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong, 666100, China.,Yunnan Key Laboratory of Southern Medicinal Utilization, Jinghong, 666100, China
| | - Xuan Ding
- Yunnan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong, 666100, China.,Yunnan Key Laboratory of Southern Medicinal Utilization, Jinghong, 666100, China
| | - Yin Yuan
- Yunnan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong, 666100, China.,Yunnan Key Laboratory of Southern Medicinal Utilization, Jinghong, 666100, China
| | - Ge Li
- Yunnan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong, 666100, China. .,Yunnan Key Laboratory of Southern Medicinal Utilization, Jinghong, 666100, China.
| | - Guang Li
- Yunnan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong, 666100, China. .,Yunnan Key Laboratory of Southern Medicinal Utilization, Jinghong, 666100, China.
| |
Collapse
|
15
|
Yang X, Yang Y, Liu K, Zhang C. Traditional Chinese medicine monomers: Targeting pulmonary artery smooth muscle cells proliferation to treat pulmonary hypertension. Heliyon 2023; 9:e14916. [PMID: 37128338 PMCID: PMC10147991 DOI: 10.1016/j.heliyon.2023.e14916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 02/01/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Pulmonary hypertension (PH) is a complex multifactorial disease characterized by increased pulmonary vascular resistance and pulmonary vascular remodeling (PVR), with high morbidity, disability, and mortality. The abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) is the main pathological change causing PVR. At present, clinical treatment drugs for PH are limited, which can only improve symptoms and reduce hospitalization but cannot delay disease progression and reduce survival rate. In recent years, numerous studies have shown that traditional Chinese medicine monomers (TCMs) inhibit excessive proliferation of PASMCs resulting in alleviating PVR through multiple channels and multiple targets, which has attracted more and more attention in the treatment of PH. In this paper, the experimental evidence of inhibiting PASMCs proliferation by TCMs was summarized to provide some directions for the future development of these mentioned TCMs as anti-PH drugs in clinical.
Collapse
|
16
|
Wang M, Tang B, Huang H, Wu X, Deng H, Chen H, Mei L, Chen X, Burgering B, Lu C. Deciphering the mechanism of PSORI-CM02 in suppressing keratinocyte proliferation through the mTOR/HK2/glycolysis axis. Front Pharmacol 2023; 14:1152347. [PMID: 37089953 PMCID: PMC10119413 DOI: 10.3389/fphar.2023.1152347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Hyperplasia of epidermal keratinocytes that depend on glycolysis is a new hallmark of psoriasis pathogenesis. Our previous studies demonstrated that PSORI-CM02 could halt the pathological progression of psoriasis by targeting inflammatory response and angiogenesis, but its effect(s) and mechanism(s) on proliferating keratinocytes remained unclear. In this study, we aim to identify components of PSORI-CM02 that are absorbed into the blood and to determine the effect(s) of PSORI-CM02 on keratinocyte proliferation and its molecular mechanism(s). We used the immortalized human epidermal keratinocyte cell line, HaCaT, as an in vitro model of proliferating keratinocytes and the imiquimod-induced psoriasis mouse (IMQ) as an in vivo model. Metabolite profiles of vehicle pharmaceutic serum (VPS), PSORI-CM02 pharmaceutic serum (PPS), and water extraction (PWE) were compared, and 23 components of PSORI-CM02 were identified that were absorbed into the blood of mice. Both PPS and PWE inhibited the proliferation of HaCaT cells and consequently reduced the expression of the proliferation marker ki67. Additionally, PPS and PWE reduced phosphorylation levels of mTOR pathway kinases. Seahorse experiments demonstrated that PPS significantly inhibited glycolysis, glycolytic capacity, and mitochondrial respiration, thus reducing ATP production in HaCaT cells. Upon treatments of PPS or PWE, hexokinase 2 (HK2) expression was significantly decreased, as observed from the set of glycolytic genes we screened. Finally, in the IMQ model, we observed that treatment with PSORI-CM02 or BPTES, an inhibitor of mTOR signaling, reduced hyperproliferation of epidermal keratinocytes, inhibited the expression of p-S6 and reduced the number of proliferating cell nuclear antigen (PCNA)-positive cells in lesioned skin. Taken together, we demonstrate that PSORI-CM02 has an anti-proliferative effect on psoriatic keratinocytes, at least in part, by inhibiting the mTOR/HK2/glycolysis axis.
Collapse
Affiliation(s)
- Maojie Wang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Molecular Cancer Research, Center of Molecular Medicine, University Medical Center Utrecht and the Oncode Institute, Utrecht, Netherlands
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bin Tang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Huanjie Huang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Molecular Cancer Research, Center of Molecular Medicine, University Medical Center Utrecht and the Oncode Institute, Utrecht, Netherlands
| | - Xiaodong Wu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Hao Deng
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Haiming Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Liyan Mei
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Xiumin Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Boudewijn Burgering
- Molecular Cancer Research, Center of Molecular Medicine, University Medical Center Utrecht and the Oncode Institute, Utrecht, Netherlands
- *Correspondence: Boudewijn Burgering, ; Chuanjian Lu,
| | - Chuanjian Lu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- *Correspondence: Boudewijn Burgering, ; Chuanjian Lu,
| |
Collapse
|
17
|
Crosstalk between xanthine oxidase (XO) inhibiting and cancer chemotherapeutic properties of comestible flavonoids- a comprehensive update. J Nutr Biochem 2022; 110:109147. [PMID: 36049673 DOI: 10.1016/j.jnutbio.2022.109147] [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: 06/03/2021] [Revised: 12/17/2021] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Gout is an inflammatory disease caused by metabolic disorder or genetic inheritance. People throughout the world are strongly dependent on ethnomedicine for the treatment of gout and some receive satisfactory curative treatment. The natural remedies as well as established drugs derived from natural sources or synthetically made exert their action by mechanisms that are closely associated with anticancer treatment mechanisms regarding inhibition of xanthine oxidase, feedback inhibition of de novo purine synthesis, depolymerization and disappearance of microtubule, inhibition of NF-ĸB activation, induction of TRAIL, promotion of apoptosis, and caspase activation and proteasome inhibition. Some anti-gout and anticancer novel compounds interact with same receptors for their action, e.g., colchicine and colchicine analogues. Dietary flavonoids, i.e., chrysin, kaempferol, quercetin, fisetin, pelargonidin, apigenin, luteolin, myricetin, isorhamnetin, phloretinetc etc. have comparable IC50 values with established anti-gout drug and effective against both cancer and gout. Moreover, a noticeable number of newer anticancer compounds have already been isolated from plants that have been using by local traditional healers and herbal practitioners to treat gout. Therefore, the anti-gout plants might have greater potentiality to become selective candidates for screening of newer anticancer leads.
Collapse
|
18
|
Wang M, Xu Z, Cai Q, Deng Y, Shi W, Zhou H, Wang D, Li J. Isorhamnetin inhibits progression of ovarian cancer by targeting ESR1. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1216. [PMID: 36544694 PMCID: PMC9761148 DOI: 10.21037/atm-22-5064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022]
Abstract
Background Although reports suggest Chinese herbal medicine treatment of ovarian cancer (OC) has a good effect, the role of isorhamnetin (ISO), a flavonol aglycone with immune, anti-inflammatory, cardiovascular and cerebrovascular protective effects, as well as an anticancer effect, in OC remains unclear. Network pharmacology was used to explore this in vitro and in vivo, and to identify relevant targets. Methods The common targets of ISO in the treatment of OC were screened by constructing drug targets and disease gene databases for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. The protein-protein interaction network was constructed by STRING. Overlapping targets were further analyzed using the online tool UALCAN to analyze the correlation between gene expression and patient survival and prognosis. The effect of ISO on OC cell proliferation, migration, and invasion was assessed in vivo and in vitro, and the function of the estrogen receptor 1 (ESR1) in the development of OC was examined by overexpressing and knocking down ESR1 expression. Results Through network pharmacology analysis, 25 target genes related to ISO-OC were screened out. The overall survival rate of OC patients only significantly correlated with high expression of ESR1 among 13 highly expressed overlapping genes. ISO significantly inhibited the proliferation, migration and invasion of OC cells in vitro and inhibited tumor growth in vivo. Overexpression of ESR1 significantly promoted the proliferation, migration and invasion of OC cells, whereas knockdown of ESR1 showed the opposite result. In addition, overexpression of ESR1 significantly reversed the inhibitory effect of ISO on the proliferation, migration and invasion of OC cells. Conclusions We confirmed that ISO inhibits OC cell proliferation, migration and invasion by targeting ESR1 expression, which provides a theoretical basis for further pharmacological research.
Collapse
Affiliation(s)
- Manman Wang
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Zhengtan Xu
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Qi Cai
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Yanmei Deng
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Weiqiao Shi
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Hongyu Zhou
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Dajiang Wang
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Jian Li
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Hernández-Caballero ME, Sierra-Ramírez JA, Villalobos-Valencia R, Seseña-Méndez E. Potential of Kalanchoe pinnata as a Cancer Treatment Adjuvant and an Epigenetic Regulator. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196425. [PMID: 36234962 PMCID: PMC9573125 DOI: 10.3390/molecules27196425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 02/05/2023]
Abstract
Cancer is a global public health problem that is related to different environmental and lifestyle factors. Although the combination of screening, prevention, and treatment of cancer has resulted in increased patient survival, conventional treatments sometimes have therapeutic limitations such as resistance to drugs or severe side effects. Oriental culture includes herbal medicine as a complementary therapy in combination with chemotherapy or radiotherapy. This study aimed to identify the bioactive ingredients in Kalanchoe pinnata, a succulent herb with ethnomedical applications for several diseases, including cancer, and reveal its anticancer mechanisms through a molecular approach. The herb contains gallic acid, caffeic acid, coumaric acid, quercetin, quercitrin, isorhamnetin, kaempferol, bersaldegenin, bryophyllin a, bryophyllin c, bryophynol, bryophyllol and bryophollone, stigmasterol, campesterol, and other elements. Its phytochemicals participate in the regulation of proliferation, apoptosis, cell migration, angiogenesis, metastasis, oxidative stress, and autophagy. They have the potential to act as epigenetic drugs by reverting the acquired epigenetic changes associated with tumor resistance to therapy-such as the promoter methylation of suppressor genes, inhibition of DNMT1 and DNMT3b activity, and HDAC regulation-through methylation, thereby regulating the expression of genes involved in the PI3K/Akt/mTOR, Nrf2/Keap1, MEK/ERK, and Wnt/β-catenin pathways. All of the data support the use of K. pinnata as an adjuvant in cancer treatment.
Collapse
Affiliation(s)
- Marta Elena Hernández-Caballero
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, Biomedicina, 13 sur 2702 Col. Volcanes, Puebla C.P. 72410, Mexico
- Correspondence: or
| | - José Alfredo Sierra-Ramírez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Díaz Mirón Esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Mexico City 11340, Mexico
| | - Ricardo Villalobos-Valencia
- UMAE Hospital de Oncología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Cd México C.P. 06725, Mexico
| | - Emmanuel Seseña-Méndez
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, Biomedicina, 13 sur 2702 Col. Volcanes, Puebla C.P. 72410, Mexico
| |
Collapse
|
20
|
Isorhamnetin Suppresses Human Gastric Cancer Cell Proliferation through Mitochondria-Dependent Apoptosis. Molecules 2022; 27:molecules27165191. [PMID: 36014431 PMCID: PMC9415531 DOI: 10.3390/molecules27165191] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Derivates of natural products have been wildly utilized in the treatment of malignant tumors. Isorhamnetin (ISO), a most important active ingredient derived from flavonoids, shows great potential in tumor therapy. However, the therapeutic effects of ISO on gastric cancer (GC) remain unclear. Here, we demonstrate that ISO treatment dramatically inhibited the proliferation of two types of GC cells (AGS-1 and HGC-27) both in vitro and in vivo in time- and dose-dependent manners. These results are consistent with the transcriptomic analysis of ISO-treated GC cells, which yielded hundreds of differentially expressed genes that were enriched with cell growth and apoptosis. Mechanically, ISO treatment initiated the activation of caspase-3 cascade and elevated the expression of mitochondria-associated Bax/Bcl-2, cytosolic cytochrome c, followed by the activation of the cleavage of caspase-3 as well as poly ADP-ribose polymerase (PARP), resulting in the severe reduction of the mitochondrial potential and the accumulation of reactive oxygen species (ROS), while pre-treatment of the caspase-3 inhibitor could block the anti-tumor effect. Therefore, these results indicate that ISO treatment induces the apoptosis of GC cells through the mitochondria-dependent apoptotic pathway, providing a potential strategy for clinical GC therapy.
Collapse
|
21
|
Fakhri S, Moradi SZ, Yarmohammadi A, Narimani F, Wallace CE, Bishayee A. Modulation of TLR/NF-κB/NLRP Signaling by Bioactive Phytocompounds: A Promising Strategy to Augment Cancer Chemotherapy and Immunotherapy. Front Oncol 2022; 12:834072. [PMID: 35299751 PMCID: PMC8921560 DOI: 10.3389/fonc.2022.834072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Background Tumors often progress to a more aggressive phenotype to resist drugs. Multiple dysregulated pathways are behind this tumor behavior which is known as cancer chemoresistance. Thus, there is an emerging need to discover pivotal signaling pathways involved in the resistance to chemotherapeutic agents and cancer immunotherapy. Reports indicate the critical role of the toll-like receptor (TLR)/nuclear factor-κB (NF-κB)/Nod-like receptor pyrin domain-containing (NLRP) pathway in cancer initiation, progression, and development. Therefore, targeting TLR/NF-κB/NLRP signaling is a promising strategy to augment cancer chemotherapy and immunotherapy and to combat chemoresistance. Considering the potential of phytochemicals in the regulation of multiple dysregulated pathways during cancer initiation, promotion, and progression, such compounds could be suitable candidates against cancer chemoresistance. Objectives This is the first comprehensive and systematic review regarding the role of phytochemicals in the mitigation of chemoresistance by regulating the TLR/NF-κB/NLRP signaling pathway in chemotherapy and immunotherapy. Methods A comprehensive and systematic review was designed based on Web of Science, PubMed, Scopus, and Cochrane electronic databases. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed to include papers on TLR/NF-κB/NLRP and chemotherapy/immunotherapy/chemoresistance by phytochemicals. Results Phytochemicals are promising multi-targeting candidates against the TLR/NF-κB/NLRP signaling pathway and interconnected mediators. Employing phenolic compounds, alkaloids, terpenoids, and sulfur compounds could be a promising strategy for managing cancer chemoresistance through the modulation of the TLR/NF-κB/NLRP signaling pathway. Novel delivery systems of phytochemicals in cancer chemotherapy/immunotherapy are also highlighted. Conclusion Targeting TLR/NF-κB/NLRP signaling with bioactive phytocompounds reverses chemoresistance and improves the outcome for chemotherapy and immunotherapy in both preclinical and clinical stages.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Akram Yarmohammadi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Narimani
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Carly E. Wallace
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| |
Collapse
|
22
|
Yuan L, Cai Y, Zhang L, Liu S, Li P, Li X. Promoting Apoptosis, a Promising Way to Treat Breast Cancer With Natural Products: A Comprehensive Review. Front Pharmacol 2022; 12:801662. [PMID: 35153757 PMCID: PMC8836889 DOI: 10.3389/fphar.2021.801662] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is one of the top-ranked malignant carcinomas associated with morbidity and mortality in women worldwide. Chemotherapy is one of the main approaches to breast cancer treatment. Breast cancer initially responds to traditional first- and second-line drugs (aromatase inhibitor, tamoxifen, and carboplatin), but eventually acquires resistance, and certain patients relapse within 5 years. Chemotherapeutic drugs also have obvious toxic effects. In recent years, natural products have been widely used in breast cancer research because of their low side effects, low toxicity, and good efficacy based on their multitarget therapy. Apoptosis, a programmed cell death, occurs as a normal and controlled process that promotes cell growth and death. Inducing apoptosis is an important strategy to control excessive breast cancer cell proliferation. Accumulating evidence has revealed that natural products become increasingly important in breast cancer treatment by suppressing cell apoptosis. In this study, we reviewed current studies on natural product–induced breast cancer cell apoptosis and summarized the proapoptosis mechanisms including mitochondrial, FasL/Fas, PI3K/AKT, reactive oxygen species, and mitogen-activated protein kinase–mediated pathway. We hope that our review can provide direction in the search for candidate drugs derived from natural products to treat breast cancer by promoting cell apoptosis.
Collapse
Affiliation(s)
- Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Liang Zhang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Sijia Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Pan Li
- Department of Pharmacy, Fengdu County Hospital of Traditional Chinese Medicine, Chongqing, China
- *Correspondence: Xiaoli Li, ; Pan Li,
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
- *Correspondence: Xiaoli Li, ; Pan Li,
| |
Collapse
|
23
|
Antunes-Ricardo M, Guardado-Félix D, Rocha-Pizaña MR, Garza-Martínez J, Acevedo-Pacheco L, Gutiérrez-Uribe JA, Villela-Castrejón J, López-Pacheco F, Serna-Saldívar SO. Opuntia ficus-indica Extract and Isorhamnetin-3-O-Glucosyl-Rhamnoside Diminish Tumor Growth of Colon Cancer Cells Xenografted in Immune-Suppressed Mice through the Activation of Apoptosis Intrinsic Pathway. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2021; 76:434-441. [PMID: 34786663 DOI: 10.1007/s11130-021-00934-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 06/13/2023]
Abstract
This study aimed to evaluate the effects of Opuntia ficus-indica extract (OFI-E) and its glycoside isorhamnetin-3-O-glucosyl-rhamnoside (IGR) on the growth of human colorectal adenocarcinoma cells and in a xenografted-immunosuppressed mice model. The IC50 values of OFI-E and IGR on colon cancer cells (HT-29 RFP) were determinate, as well as their effects on the cell cycle and apoptosis induction. OFI-E and IGR produced an increased in apoptosis induction, ROS production and a G0/G1 cell cycle arrest. In xenografted-inmunosupressed mice, OFI-E and IGR reduced the tumor growth rate, myeloperoxidase activity and total cholesterol levels. OFI-E and IGR reduced the tumor growth through the overexpression of cleaved Caspase-9, Hdac11, and Bai1 proteins. OFI-E reduced the expression of bcl-2. Results demonstrated the chemopreventive effects of OFI-E, and its purified compound IGR, showing their potential as an alternative in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- M Antunes-Ricardo
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico
| | - D Guardado-Félix
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico
| | - M R Rocha-Pizaña
- Tecnologico de Monterrey, Campus Puebla, Vía Atlixcáyotl 2301, C.P. 72453, Puebla, Puebla, Mexico
| | - J Garza-Martínez
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico
| | - L Acevedo-Pacheco
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico
| | - J A Gutiérrez-Uribe
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico.
- Tecnologico de Monterrey, Campus Puebla, Vía Atlixcáyotl 2301, C.P. 72453, Puebla, Puebla, Mexico.
| | - J Villela-Castrejón
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico
| | - F López-Pacheco
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico
| | - S O Serna-Saldívar
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849, Monterrey, N.L., Mexico.
| |
Collapse
|
24
|
Patel P, Shah J. Protective effects of hesperidin through attenuation of Ki67 expression against DMBA-induced breast cancer in female rats. Life Sci 2021; 285:119957. [PMID: 34530017 DOI: 10.1016/j.lfs.2021.119957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 08/31/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
AIMS Doxorubicin (Dox) is routinely used for breast cancer treatment but toxicity and drug resistance limit its use. The objective of the study was to investigate the protective effects of hesperidin alone and in combination with doxorubicin against experimentally induced breast cancer in female rats. METHODS Breast cancer (BC) was induced by administration of 7,12-dimethylbenz(a)anthracene (DMBA) through subcutaneous injection into the 3rd right mammary gland of female Wistar rats. Hesperidin (Hes) pretreated groups were started with Hes (200 mg/kg) two weeks prior to DMBA induction. Animals were randomly divided into nine groups namely vehicle control, DMBA-induced, Dox 4 mg/kg, Dox 2 mg/kg, Hes (200 mg/kg), Hes (200 mg/kg) plus Dox 4 mg/kg treated groups and Hes pretreated groups treated with DMBA, Dox 4 mg/kg and Dox 2 mg/kg. KEY FINDINGS Hes pretreated groups showed reduced tumor occurrence, tumor volume and increased survival rate as compared to DMBA-induced group of animals. Hes pretreated animals treated with Dox 4 mg/kg and 2 mg/kg exhibited significant reduction in malondialdehyde and improvement in levels of glutathione and inflammatory markers like IL-6, TNF-α, NF-κB, IFN-γ as compared to Dox 4 mg/kg and 2 mg/kg treated animals. Histopathology and Ki67 expression depicted better control of tumor with Hes pretreatment groups as compared to DMBA-induced. Histopathology of vital organs of Hes pretreated groups treated with Dox revealed lesser toxicity than Dox treated groups. SIGNIFICANCE Hesperidin possesses protective effect against experimentally induced breast cancer in female rats that appears to be related to attenuation of Ki67 expression.
Collapse
Affiliation(s)
- Pankti Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Jigna Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India.
| |
Collapse
|
25
|
Ferreira-Santos P, Badim H, Salvador ÂC, Silvestre AJD, Santos SAO, Rocha SM, Sousa AM, Pereira MO, Wilson CP, Rocha CMR, Teixeira JA, Botelho CM. Chemical Characterization of Sambucus nigra L. Flowers Aqueous Extract and Its Biological Implications. Biomolecules 2021; 11:biom11081222. [PMID: 34439888 PMCID: PMC8391949 DOI: 10.3390/biom11081222] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 02/07/2023] Open
Abstract
The main goal of this study was to chemically characterize an aqueous S. nigra flower extract and validate it as a bioactive agent. The elderflower aqueous extraction was performed at different temperatures (50, 70 and 90 °C). The extract obtained at 90 °C exhibited the highest phenolic content and antiradical activity. Therefore, this extract was analyzed by GC-MS and HPLC-MS, which allowed the identification of 46 compounds, being quercetin and chlorogenic acid derivatives representative of 86% of the total of phenolic compounds identified in hydrophilic fraction of the aqueous extract. Naringenin (27.2%) was the major compound present in the lipophilic fraction. The antiproliferative effects of the S. nigra extract were evaluated using the colon cancer cell lines RKO, HCT-116, Caco-2 and the extract’s antigenotoxic potential was evaluated by the Comet assay in RKO cells. The RKO cells were the most susceptible to S. nigra flower extract (IC50 = 1250 µg mL−1). Moreover, the extract showed antimicrobial activity against Gram-positive bacteria, particularly Staphylococcus aureus and S. epidermidis. These results show that S. nigra-based extracts can be an important dietary source of bioactive phenolic compounds that contribute to health-span improving life quality, demonstrating their potential as nutraceutical, functional foods and/or cosmetic components for therapeutic purposes.
Collapse
Affiliation(s)
- Pedro Ferreira-Santos
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (H.B.); (A.M.S.); (M.O.P.); (C.P.W.); (C.M.R.R.); (J.A.T.)
- Correspondence: (P.F.-S.); (C.M.B.)
| | - Helder Badim
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (H.B.); (A.M.S.); (M.O.P.); (C.P.W.); (C.M.R.R.); (J.A.T.)
| | - Ângelo C. Salvador
- CICECO—Aveiro Institute of Materials, Chemistry Department, Campus de Santiago, University of Aveiro, 3810-1930 Aveiro, Portugal; (Â.C.S.); (A.J.D.S.); (S.A.O.S.)
| | - Armando J. D. Silvestre
- CICECO—Aveiro Institute of Materials, Chemistry Department, Campus de Santiago, University of Aveiro, 3810-1930 Aveiro, Portugal; (Â.C.S.); (A.J.D.S.); (S.A.O.S.)
| | - Sónia A. O. Santos
- CICECO—Aveiro Institute of Materials, Chemistry Department, Campus de Santiago, University of Aveiro, 3810-1930 Aveiro, Portugal; (Â.C.S.); (A.J.D.S.); (S.A.O.S.)
| | - Sílvia M. Rocha
- Departamento de Química & LAQV-REQUIMTE, Universidade de Aveiro, 3810-193 Aveiro, Portugal;
| | - Ana M. Sousa
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (H.B.); (A.M.S.); (M.O.P.); (C.P.W.); (C.M.R.R.); (J.A.T.)
| | - Maria Olívia Pereira
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (H.B.); (A.M.S.); (M.O.P.); (C.P.W.); (C.M.R.R.); (J.A.T.)
| | - Cristina Pereira Wilson
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (H.B.); (A.M.S.); (M.O.P.); (C.P.W.); (C.M.R.R.); (J.A.T.)
- Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Cristina M. R. Rocha
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (H.B.); (A.M.S.); (M.O.P.); (C.P.W.); (C.M.R.R.); (J.A.T.)
| | - José António Teixeira
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (H.B.); (A.M.S.); (M.O.P.); (C.P.W.); (C.M.R.R.); (J.A.T.)
| | - Cláudia M. Botelho
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (H.B.); (A.M.S.); (M.O.P.); (C.P.W.); (C.M.R.R.); (J.A.T.)
- Correspondence: (P.F.-S.); (C.M.B.)
| |
Collapse
|
26
|
Li X, Chen H, Zhang Z, Xu D, Duan J, Li X, Yang L, Hua R, Cheng J, Li Q. Isorhamnetin Promotes Estrogen Biosynthesis and Proliferation in Porcine Granulosa Cells via the PI3K/Akt Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:6535-6542. [PMID: 34096286 DOI: 10.1021/acs.jafc.1c01543] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Isorhamnetin is a natural flavonoid widely distributed in fruits and vegetables. However, the roles of isorhamnetin involved in steroidogenesis, proliferation, and apoptosis in ovarian granulosa cells (GCs) are poorly understood. We found that isorhamnetin promoted the secretion of estrogen and inhibited the secretion of progesterone and testosterone by modulating steroidogenesis-associated proteins and mRNA such as CYP19A1, StAR, and 3β-HSD in ovarian GCs. Mechanistically, isorhamnetin stimulated the expression of the proliferating cell nuclear antigen and C-myc and promoted the proliferation of GCs via the PI3K/Akt signaling pathway. Furthermore, isorhamnetin increased the protein expression of CyclinB, CyclinD, CyclinE, and CyclinA, thereby raising the ratio of S-phase cells in response to GC proliferation. Changes in the expression of apoptosis-associated proteins (Bcl2, Bax, and cytochrome c) and intracellular reactive oxygen species levels showed that isorhamnetin inhibited GC apoptosis. Collectively, these findings indicate that isorhamnetin regulates steroidogenesis through the activation of PI3K/Akt, which promotes proliferation, inhibits apoptosis, and alleviates oxidative stress.
Collapse
Affiliation(s)
- Xiaoya Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, P. R. China
| | - Huali Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621000, P. R. China
| | - Zelin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, P. R. China
| | - Dejun Xu
- College of Animal Science and Technology, Southwest University, Chongqing 400000, P. R. China
| | - Jiaxin Duan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, P. R. China
| | - Xiaodi Li
- College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050000, P. R.China
| | - Li Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, P. R. China
| | - Rongmao Hua
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, P. R. China
| | - Jianyong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, P. R. China
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, P. R. China
| |
Collapse
|
27
|
Isorhamnetin inhibited the proliferation and metastasis of androgen-independent prostate cancer cells by targeting the mitochondrion-dependent intrinsic apoptotic and PI3K/Akt/mTOR pathway. Biosci Rep 2021; 40:222067. [PMID: 32039440 PMCID: PMC7080645 DOI: 10.1042/bsr20192826] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/05/2019] [Accepted: 02/05/2020] [Indexed: 12/15/2022] Open
Abstract
The present study investigated the effects of Isorhamnetin on two types of prostate cancer cells (androgen-independent and androgen-dependent) and explored its possible mechanisms underlying such effects. Treatment with Isorhamnetin significantly inhibited cell growth and induced lactate dehydrogenase (LDH) release of androgen-independent DU145 and PC3 prostate cancer cells, but exhibited almost no toxicity effect on androgen-dependent LNCaP prostate cancer cell line or normal human prostate epithelial PrEC cells, which was achieved by the induction of apoptosis in a mitochondrion-dependent intrinsic apoptotic pathway. Furthermore, Isorhamnetin inhibited cell migration and invasion in concentration-dependent manners by enhancing mesenchymal−epithelial transition (MET) and inhibiting matrix metalloproteinase (MMP) 2 (MMP-2) and MMP-9 overexpression. In addition, Isorhamnetin also down-regulated the expression of phosphorylated PI3K (p-P13K), Akt (p-Akt), and mTOR (p-mTOR) proteins in both cancer cells, revealing Isorhamnetin to be a selective PI3K–Akt–mTOR pathway inhibitor. In summary, these findings propose that Isorhamnetin might be a novel therapeutic candidate for the treatment of androgen-independent prostate cancer.
Collapse
|
28
|
Uchida Y, Ferdousi F, Zheng YW, Oda T, Isoda H. Global Gene Expression Profiling Reveals Isorhamnetin Induces Hepatic-Lineage Specific Differentiation in Human Amniotic Epithelial Cells. Front Cell Dev Biol 2020; 8:578036. [PMID: 33224947 PMCID: PMC7674172 DOI: 10.3389/fcell.2020.578036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
Human amnion epithelial cells (hAECs), derived from discarded term placenta, is anticipated as a new stem cell resource because of their advantages over embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), such as no risk of tumorigenicity and minimal ethical issue. hAECs have been reported to differentiate into hepatic-like cells (HLCs) with variable functionalities suitable for cell-based therapy of end-stage liver diseases, drug screening, and drug toxicity tests. On the other hand, a new research stream has been evolving to use natural compounds as stimulants of stem cell differentiation because of their high availability and minimum side effects. Isorhamnetin is a naturally occurring flavonoid commonly found in fruits and vegetables and has been reported to improve hepatic fibrosis and steatosis. In this present study, we have screened the differentiation potential of isorhamnetin in hAECs. The cells were grown on 3D cell culture and were treated with 20 μM of synthesized isorhamnetin for 10 days without adding any additional growth factors. DNA microarray global gene expression analysis was conducted for differentially expressed genes between isorhamnetin-treated and untreated control cells, gene expression validation was carried out using RT-qPCR method, and finally, several hepatic functions were assessed. Microarray analysis showed that isorhamnetin could activate essential biological processes, molecular functions, and signaling pathways for hepatic differentiation. Hepatic progenitor markers, EPCAM and DLK1, were upregulated in the isorhamnetin-treated hAECs. AFP was downregulated, while ALB was upregulated on Day 10. Furthermore, isorhamnetin-treated cells could show increased CYP enzyme mRNA levels, ICG uptake and release, glycogen storage activity, and urea secretion. Additionally, isorhamnetin-treated cells did not show any trace of transdifferentiation evident by significant downregulation of several colon- and cholangiocyte-specific markers. However, longer treatment with isorhamnetin did not promote hepatic maturation. Altogether, our findings indicate that isorhamnetin has a promising effect on directing the hepatic-lineage specific differentiation in hAECs.
Collapse
Affiliation(s)
- Yoshiaki Uchida
- School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Japan
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | - Yun-Wen Zheng
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tatsuya Oda
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan.,Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroko Isoda
- School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Japan.,Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
29
|
Aonuma K, Ferdousi F, Xu D, Tominaga K, Isoda H. Effects of Isorhamnetin in Human Amniotic Epithelial Stem Cells in vitro and Its Cardioprotective Effects in vivo. Front Cell Dev Biol 2020; 8:578197. [PMID: 33117805 PMCID: PMC7552739 DOI: 10.3389/fcell.2020.578197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiac hypertrophy and fibrosis are major pathophysiologic disorders that lead to serious cardiovascular diseases (CVDs), such as heart failure and arrhythmia. It is well known that transforming growth factor β (TGFβ) signaling pathways play a major role in the proliferation of cardiac hypertrophy and fibrosis, which is mainly stimulated by angiotensin II (AgII). This study aimed to investigate the cardioprotective potential of isorhamnetin (ISO) in human amniotic epithelial stem cells (hAESCs) through global gene expression analysis and to confirm its beneficial effects on cardiac hypertrophy and fibrosis in the AgII-induced in vivo model. In vitro, biological processes including TGFβ, collagen-related functions, and inflammatory processes were significantly suppressed in ISO pretreated hAESCs. In vivo, continuous AgII infusion using an osmotic pump induced significant pathological fibrosis and myocardial hypertrophy, which were remarkably suppressed by ISO pretreatment. ISO was found to reverse the enhanced TGFβ and Collagen type I alpha 1 mRNA expression induced by AgII exposure, which causes cardiovascular remodeling in ventricular tissue. These findings indicate that ISO could be a potential agent against cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Kazuhiro Aonuma
- School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Japan
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | - Farhana Ferdousi
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
| | - DongZhu Xu
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
- Cardiovascular Division, Institute of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kenichi Tominaga
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | - Hiroko Isoda
- School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Japan
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
30
|
Chen Q, Song S, Wang Z, Shen Y, Xie L, Li J, Jiang L, Zhao H, Feng X, Zhou Y, Zhou M, Zeng X, Ji N, Chen Q. Isorhamnetin induces the paraptotic cell death through ROS and the ERK/MAPK pathway in OSCC cells. Oral Dis 2020; 27:240-250. [PMID: 32654232 DOI: 10.1111/odi.13548] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE There were rarely investigations on the effects and molecular mechanisms of oral squamous cell carcinoma (OSCC) cells when treated with isorhamnetin. This article assesses the anti-cancer effect of isorhamnetin. METHODS AND MATERIALS Oral squamous cell carcinoma cells were treated with or without isorhamnetin. Cell proliferation, cell cycle arrest, cell migration, cell death, and the related signaling pathways were evaluated. RESULTS The results revealed that cell proliferation was inhibited in a dose- and time-dependent manner, which was confirmed by diminished cell viability and revealed by decreased in the number of cell colonies. In addition, the cell cycle arrested in the G2/M phase, and the protein levels of cyclin B1 and CDC2 were suppressed. Moreover, the cell migration was inhibited, and the protein levels of related proteins were modulated. Furthermore, it could be observed that abundant cytoplasmic vacuoles existed which that were derived from mitochondria and the endoplasmic reticulum. It was confirmed that cell death did not result from apoptosis and may have which may be apt to paraptosis. Isorhamnetin was observed to upregulate phosphorylated ERK cascades and increase intracellular reactive oxygen species levels. CONCLUSIONS Our study suggested that the anti-cancer effect of isorhamnetin might trigger paraptosis, which may indicate a new therapeutic approach to OSCC.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Shaojuan Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Zhen Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Yingqiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Xiaodong Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Yu Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Min Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Systems Pharmacology-Based Research on the Mechanism of Tusizi-Sangjisheng Herb Pair in the Treatment of Threatened Abortion. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4748264. [PMID: 32775426 PMCID: PMC7391104 DOI: 10.1155/2020/4748264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/03/2020] [Indexed: 12/22/2022]
Abstract
Threatened abortion (TA) is a common complication with high incidence in the first trimester of pregnancy, which will end in miscarriage if not treated properly. The Chinese herbs Cuscutae Semen (Tusizi in Chinese) and Herba Taxilli (Sangjisheng in Chinese) first recorded in the ancient classic medical book Shennong Bencao Jing are effective and widely used as an herb pair for the treatment of TA, while the active ingredients and the functional mechanism of Tusizi-Sangjisheng herb pair treating TA are still unknown. In order to exploit the relationship between those two herbs and TA, systems pharmacology analysis was carried out in this study. A total of 75 ingredients of Tusizi-Sangjisheng were collected from Traditional Chinese Medicine System Pharmacology Database and Analysis Platform (TCMSP). 12 bioactive compounds were screened, and 153 directly related targets were predicted by systematic models. Besides, Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were used to systematically explore the potential mechanisms of Tusizi-Sangjisheng treating TA. Meanwhile, Compound-Target (C-T), Target-Disease (T-D), and Target-Pathway (T-P) networks were constructed to further quest the underlying functional mechanisms of Tusizi-Sangjisheng. As a result, 31 targets and 3 key pathways were found to be directly related to TA that includes mitogen-activated protein kinases (MAPKs), phosphatidylinositol-3-kinase/protein kinase B (PI3K-Akt), and transforming growth factor-β (TGF-β) signaling pathways. The results in this study may provide some valuable clues about the molecular mechanisms of the efficient Chinese herb pair Tusizi-Sangjisheng in the treatment of TA.
Collapse
|
32
|
Exploring Mechanism of Key Chinese Herbal Medicine on Breast Cancer by Data Mining and Network Pharmacology Methods. Chin J Integr Med 2020; 27:919-926. [PMID: 32572780 DOI: 10.1007/s11655-020-3422-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To screen the key Chinese Herbal Medicines (KCHMs) against breast cancer by data mining, and analyze the potential mechanism of KCHMs using network pharmacology method. METHODS Clinical prescriptions consisted of CHMs for treating breast cancer were screened, and then Traditional Chinese Medicine Inheritance Support System (TCMISS) was applied to obtain the KCHMs. Subsequently, active ingredients and corresponding target genes of KCHMs were searched by Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) database, and target genes of breast cancer were collected using OMIM and MalaCards. After that, the overlapping target genes of KCHMs and breast cancer were screened, and the protein-protein interaction (PPI) network was built. In addition, a network of "KCHMs-active ingredients-breast cancer-targets" was constructed by Cytoscape 3.7.1. Finally, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis were performed with Database for Annotation, Visualization and Integrated Discovery (DAVID) database to reveal the action mechanism of KCHMs. RESULTS A total of 7 KCHMs were identified, whose active ingredients include quercetin, luteolin, nobiletin, kaempferol, isorhamnetin, naringenin, and be-ta-sitosterol, etc. Based on protein-protein interaction analysis, core targets were ESR1, MYC, CCND1, EGFR, CASP3, ERBB2, etc. Several KEGG pathways (e.g, PI3K-Akt, p53, ErbB, and HIF-1 signaling pathways) were found. CONCLUSION Based on the combination of the data mining method and network pharmacology approach, the therapeutic effect of KCHMs on breast cancer may be realized by acting on target genes and signaling pathways related to the formation and progression of breast cancer.
Collapse
|
33
|
Rahemi S, Nematollahi-Mahani SN, Rajaie A, Fallah H. Inhibitor of Interleukin-1 Receptor-associated Kinases 1/4, Can Increase the Sensitivity of Breast Cancer Cells to Methotrexate. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2020; 8:200-209. [PMID: 32489949 PMCID: PMC7241845 DOI: 10.22088/ijmcm.bums.8.3.200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Breast cancer is the most common type of cancer among women. Chemotherapy is one of the main methods of breast cancer treatment, but this method is increasingly affected due to drug resistance. One of the newly discovered factors associated with drug resistance in cancer cells is interleukin receptor-associated kinase 1 (IRAK1). The aim of this study was to investigate the relationship between IRAK1 inhibition and sensitivity to methotrexate (MTX). Effects of various concentrations of MTX and constant concentration (1μg/ml) of IRAK1/4 inhibitor was examined on MCF-7, BT-20, BT-549, MB-468 cell lines. Cell viability was examined by water soluble tetrazole -1, and cell apoptosis by flow cytometry. The expression of IRAK1 and BCRP genes was also assessed by real-time PCR method. IRAK1 inhibitor decreased IC50 in all examined cell lines, but the most prominent effect was observed in MB-468. 72 h incubation of cell lines with IRAK inhibitor and MTX, significantly increased the annexin-V and annexin-V/7AAD positive cells, suggesting an apoptotic effect of IRAK on all examined breast cancer cell lines. RT-qPCR test results showed that the IRAK inhibitor had no effect on the expression of BCRP at any time. Our results showed that IRAK inhibitor can increase the chemosensitivity of breast cancer cell lines without effect on BCRP mRNA expression. IRAK inhibitor in combination with MTX can induce apoptosis in breast cancer cell lines.
Collapse
Affiliation(s)
- Samaneh Rahemi
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Athena Rajaie
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Fallah
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
34
|
Gong G, Guan YY, Zhang ZL, Rahman K, Wang SJ, Zhou S, Luan X, Zhang H. Isorhamnetin: A review of pharmacological effects. Biomed Pharmacother 2020; 128:110301. [PMID: 32502837 DOI: 10.1016/j.biopha.2020.110301] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/11/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
Isorhamnetin is one of the most important active ingredients in the fruits of Hippophae rhamnoides L. and the leaves of Ginkgo biloba L., which possesses extensive pharmacological activities. At present, there have been numerous investigations on isorhamnetin, which has the effects of cardiovascular and cerebrovascular protection, anti-tumor, anti-inflammatory, anti-oxidation, organ protection, prevention of obesity, etc. The related mechanisms involve the regulation of PI3K/AKT/PKB, NF-κB, MAPK and other signaling pathways as well as the expression of related cytokines and kinases. Isorhamnetin has a high value of development and application. However, the investigations on its mechanism of action are limited and lack of detailed scientific validation. The manuscript reviewed the pharmacological effects of isorhamnetin and related mechanisms of action for the development of its medicinal properties further.
Collapse
Affiliation(s)
- Gang Gong
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Ying-Yun Guan
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhong-Lin Zhang
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, PR China
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, L3 3AF, England, UK
| | - Su-Juan Wang
- Department of Drug Preparation, Hospital of TCM and Hui Nationality Medicine, Ningxia Medical University, Wuzhong, PR China
| | - Shuang Zhou
- Acupuncture and Moxibustion Techniques Department, School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai, 200032, PR China.
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| | - Hong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| |
Collapse
|
35
|
Zhang J, Li X, Huang L. Anticancer activities of phytoconstituents and their liposomal targeting strategies against tumor cells and the microenvironment. Adv Drug Deliv Rev 2020; 154-155:245-273. [PMID: 32473991 PMCID: PMC7704676 DOI: 10.1016/j.addr.2020.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/07/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
Various bioactive ingredients have been extracted from Chinese herbal medicines (CHMs) that affect tumor progression and metastasis. To further understand the mechanisms of CHMs in cancer therapy, this article summarizes the effects of five categories of CHMs and their active ingredients on tumor cells and the tumor microenvironment. Despite their treatment potential, the undesirable physicochemical properties (poor permeability, instability, high hydrophilicity or hydrophobicity, toxicity) and unwanted pharmacokinetic profiles (short half-life in blood and low bioavailability) restrict clinical studies of CHMs. Therefore, development of liposomes through relevant surface modifying techniques to achieve targeted CHM delivery for cancer cells, i.e., extracellular and intracellular targets and targets in tumor microenvironment or vasculature, have been reviewed. Current challenges of liposomal targeting of these phytoconstituents and future perspective of CHM applications are discussed to provide an informative reference for interested readers.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Xiang Li
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
36
|
Chang Z, Wang JL, Jing ZC, Ma P, Xu QB, Na JR, Tian J, Ma X, Zhou W, Zhou R. Protective effects of isorhamnetin on pulmonary arterial hypertension: in vivo and in vitro studies. Phytother Res 2020; 34:2730-2744. [PMID: 32452118 DOI: 10.1002/ptr.6714] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/15/2020] [Accepted: 04/18/2020] [Indexed: 12/14/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a malignant disease with high mortality and closely involves the bone morphogenetic protein (BMP) pathway. Mutations in BMPR2 caused proliferation of pulmonary artery smooth muscle cells (PASMCs) leading to PAH. Isorhamnetin, one of the main naturally occurring flavonoids extracted from Hippophae rhamnoides L, shows antiinflammatory and anti-proliferative properties. Nevertheless, the effects of isorhamnetin on PAH remain unclear. This study aimed to investigate whether isorhamnetin has protective effects against PAH and explore possible mechanisms. An in vivo model of PAH induced by monocrotaline (MCT) was employed, and sildenafil and isorhamnetin were orally administered for 21 consecutive days. An in vitro model induced by TNF-α was employed, and cell proliferation of HPASMCs was detected. Results indicated that isorhamnetin significantly improved hemodynamic, histopathological, and echocardiographic changes in MCT-induced PAH in rats. In vitro, isorhamnetin suppressed TNF-α-induced HPASMCs proliferation. Furthermore, isorhamnetin improved protein expression of BMPR2 and suppressed protein expression of TNF-α and IL-6 in rat lungs. Isorhamnetin improved protein expression of BMPR2 and p-smad1/5 and mRNA expression of Id1 and Id3 in HPASMCs. Isorhamnetin ameliorated MCT-induced PAH in rats and inhibited TNF-α-induced HPASMCs proliferation by a mechanism likely involving the regulation of the BMP signaling pathway.
Collapse
Affiliation(s)
- Zhi Chang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jia-Ling Wang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Zhi-Cheng Jing
- Department of Cardiology, Peking Union Medical College Hospital, Key Lab of Pulmonary Vascular Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ping Ma
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Qing-Bing Xu
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jian-Rong Na
- Respiratory and critical care medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jie Tian
- Respiratory and critical care medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xuan Ma
- Respiratory and critical care medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wei Zhou
- Respiratory and critical care medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ru Zhou
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China.,Ningxia Characteristic Traditional Chinese Medicine Modernization Engineering Technology Research Center, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
37
|
Isorhamnetin Induces Melanoma Cell Apoptosis via the PI3K/Akt and NF- κB Pathways. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1057943. [PMID: 32461960 PMCID: PMC7225865 DOI: 10.1155/2020/1057943] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/31/2020] [Indexed: 01/04/2023]
Abstract
Malignant melanoma is characterized by its bad prognosis for aggressiveness, drug resistance, and early metastasis. Isorhamnetin (3′-methoxy-3,4′,5,7-tetrahydroxyflavone; IH) is a natural flavonoid that has been investigated for its antitumor effects in breast cancer, colon cancer, and gastric cancer through inducing cell apoptosis. Given its role in tumor inhibition, no research has been conducted concerning its effect against melanoma. In the present study, we found that IH could significantly inhibit B16F10 cell proliferation and migration and induce B16F10 cell apoptosis. The examination on molecular mechanism revealed that IH could suppress the phosphorylation of Akt and the translocation of NF-κB, which are key factors in apoptosis-related pathways. We also detected that this process was related to the bifunctional 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases 4 (PFKFB4) by PFKFB4 knockdown experiment. In line with in vitro study, we further provided that IH effectively inhibited tumor growth in vivo. Taken together, IH was proven to induce melanoma cell apoptosis in vitro and in vivo, which may serve as a potential agent in malignant melanoma treatment in the future.
Collapse
|
38
|
Fan S, Yang G, Zhang J, Li J, Bai B. Optimization of Ultrasound-Assisted Extraction Using Response Surface Methodology for Simultaneous Quantitation of Six Flavonoids in Flos Sophorae Immaturus and Antioxidant Activity. Molecules 2020; 25:molecules25081767. [PMID: 32290627 PMCID: PMC7221660 DOI: 10.3390/molecules25081767] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Ultrasound-assisted extraction (UAE) was applied to extract rutin (RU), nicotiflorin (NI), narcissoside (NA), kaempferol (KA), isorhamnetin (IS), quercetin (QU), and total flavonoids of Flos Sophorae Immaturus (TFFSI) from Flos Sophorae Immaturus (FSI). Through single factor test and response surface methodology (RSM), the optimal extraction conditions were concluded as follows: ethanol concentration 70%, time 30 min, temperature 61 °C, and liquid/solid ratio 15.30 mL/g, respectively. The actual extraction rates of RU, NI, NA, KA, IS, QU, and TFFSI were 14.6101%, 2.9310%, 7.1987%, 0.1041%, 0.4920%, 2.7998%, and 26.4260%, respectively. The experimental results demonstrated that the extraction method with accuracy and efficiency could be used for the comprehensive evaluation quality control of extracts from FSI. The antioxidant activities of hydroalcoholic extraction from FSI on 1,1-diphenyl-2-picrylhydrazyl (DPPH), 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS•+), superoxide anion (•O2−) free radicals, and ferric reducing/antioxidant power (FRAP) were assessed. The results showed that the antioxidation activities of extracts on DPPH, ABTS•+, and •O2− free radicals were reached 89.29%, 97.86%, and 56.61%, and 81.4% in FRAP at 1.0 mg/mL, respectively. The antioxidant capacity of FSI extract was positively correlated with the amount of total flavonoids.
Collapse
Affiliation(s)
- Sanhong Fan
- College of Life Science, Shanxi University, Taiyuan 030000, China; (G.Y.)
- Shanxi Key Laboratory for Research and Development of Regional Plants, Taiyuan 030000, China
- Correspondence: (S.F.); (B.B.); Tel.: +86-13653644479 (S.F.); 86+15034132105 (B.B.)
| | - Gege Yang
- College of Life Science, Shanxi University, Taiyuan 030000, China; (G.Y.)
| | - Jinhua Zhang
- College of Life Science, Shanxi University, Taiyuan 030000, China; (G.Y.)
- Shanxi Key Laboratory for Research and Development of Regional Plants, Taiyuan 030000, China
| | - Jiani Li
- College of Life Science, Shanxi University, Taiyuan 030000, China; (G.Y.)
| | - Baoqing Bai
- College of Life Science, Shanxi University, Taiyuan 030000, China; (G.Y.)
- Shanxi Key Laboratory for Research and Development of Regional Plants, Taiyuan 030000, China
- Correspondence: (S.F.); (B.B.); Tel.: +86-13653644479 (S.F.); 86+15034132105 (B.B.)
| |
Collapse
|
39
|
Ganbold M, Owada Y, Ozawa Y, Shimamoto Y, Ferdousi F, Tominaga K, Zheng YW, Ohkohchi N, Isoda H. Isorhamnetin Alleviates Steatosis and Fibrosis in Mice with Nonalcoholic Steatohepatitis. Sci Rep 2019; 9:16210. [PMID: 31700054 PMCID: PMC6838085 DOI: 10.1038/s41598-019-52736-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the most severe and progressive form of nonalcoholic fatty liver disease (NAFLD), which can lead to life-threatening conditions, however, there is still no approved drug for the treatment of NASH. In this study we used human-like NASH mouse model and treated orally with isorhamnetin at a dose of 50 mg/kg to analyze the effect of isorhamnetin on the progression of NASH. NASH-induced mice represented severe steatosis with inflammation, and fibrosis in liver accompanied with high level of liver injury markers in serum. Isorhamnetin treatment reduced intrahepatic lipid accumulation and TG content by inhibiting de novo lipogenic pathway in NASH-induced mice. Consistent with this, isorhamnetin-treated NASH mice showed improved liver injury markers, reduced collagen deposition as well as decreased gene expression of fibrogenic markers. Taken together, here we showed for the first time that synthesized isorhamnetin alleviates pathologic features of NASH and thus can potentially contribute to NASH drug development.
Collapse
Affiliation(s)
- Munkhzul Ganbold
- School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan
| | - Yohei Owada
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yusuke Ozawa
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yasuhiro Shimamoto
- Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8565, Japan
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| | - Kenichi Tominaga
- Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8565, Japan
| | - Yun-Wen Zheng
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Nobuhiro Ohkohchi
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hiroko Isoda
- School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan. .,Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8565, Japan. .,Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan. .,Faculty of Life and Environment Science, University of Tsukuba, Tsukuba, 305-8572, Japan.
| |
Collapse
|
40
|
Park C, Cha HJ, Choi EO, Lee H, Hwang-Bo H, Ji SY, Kim MY, Kim SY, Hong SH, Cheong J, Kim GY, Yun SJ, Hwang HJ, Kim WJ, Choi YH. Isorhamnetin Induces Cell Cycle Arrest and Apoptosis Via Reactive Oxygen Species-Mediated AMP-Activated Protein Kinase Signaling Pathway Activation in Human Bladder Cancer Cells. Cancers (Basel) 2019; 11:cancers11101494. [PMID: 31590241 PMCID: PMC6826535 DOI: 10.3390/cancers11101494] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022] Open
Abstract
Isorhamnetin is an O-methylated flavonol that is predominantly found in the fruits and leaves of various plants, which have been used for traditional herbal remedies. Although several previous studies have reported that this flavonol has diverse health-promoting effects, evidence is still lacking for the underlying molecular mechanism of its anti-cancer efficacy. In this study, we examined the anti-proliferative effect of isorhamnetin on human bladder cancer cells and found that isorhamnetin triggered the gap 2/ mitosis (G2/M) phase cell arrest and apoptosis. Our data showed that isorhamnetin decreased the expression of Wee1 and cyclin B1, but increased the expression of cyclin-dependent kinase (Cdk) inhibitor p21WAF1/CIP1, and increased p21 was bound to Cdk1. In addition, isorhamnetin-induced apoptosis was associated with the increased expression of the Fas/Fas ligand, reduced ratio of B-cell lymphoma 2 (Bcl-2)/Bcl-2 associated X protein (Bax) expression, cytosolic release of cytochrome c, and activation of caspases. Moreover, isorhamnetin inactivated the adenosine 5′-monophosphate-activated protein kinase (AMPK) signaling pathway by diminishing the adenosine triphosphate (ATP) production due to impaired mitochondrial function. Furthermore, isorhamnetin stimulated production of intracellular reactive oxygen species (ROS); however, the interruption of ROS generation using a ROS scavenger led to an escape from isorhamnetin-mediated G2/M arrest and apoptosis. Collectively, this is the first report to show that isorhamnetin inhibited the proliferation of human bladder cancer cells by ROS-dependent arrest of the cell cycle at the G2/M phase and induction of apoptosis. Therefore, our results provide an important basis for the interpretation of the anti-cancer mechanism of isorhamnetin in bladder cancer cells and support the rationale for the need to evaluate more precise molecular mechanisms and in vivo anti-cancer properties.
Collapse
Affiliation(s)
- Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dong-eui University, Busan 47340, Korea;
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan 49267, Korea;
| | - Eun Ok Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Hyun Hwang-Bo
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea;
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea;
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - So Young Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Su Hyun Hong
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - JaeHun Cheong
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea;
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea;
| | - Seok Joong Yun
- Department of Urology, College of Medicine, Chungbuk National University, Chungbuk 8644, Korea;
| | - Hye Jin Hwang
- Department of Food and Nutrition, College of Nursing, Healthcare Sciences & Human Ecology, Dong-Eui University, Busan 47340, Korea;
| | - Wun-Jae Kim
- Department of Urology, College of Medicine, Chungbuk National University, Chungbuk 8644, Korea;
- Correspondence: (W.-J.K.); (Y.H.C.); Tel.: +82-43-269-6136 (W.-J.K.); +82-51-850-7413 (Y.H.C.)
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea; (E.O.C.); (H.L.); (H.H.-B.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
- Correspondence: (W.-J.K.); (Y.H.C.); Tel.: +82-43-269-6136 (W.-J.K.); +82-51-850-7413 (Y.H.C.)
| |
Collapse
|
41
|
Antifibrotic effect of methylated quercetin derivatives on TGFβ-induced hepatic stellate cells. Biochem Biophys Rep 2019; 20:100678. [PMID: 31467991 PMCID: PMC6711851 DOI: 10.1016/j.bbrep.2019.100678] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/02/2019] [Accepted: 08/10/2019] [Indexed: 12/18/2022] Open
Abstract
Quercetin (QCT) and isorhamnetin (ISO), natural flavonoids, were both shown to possess antifibrotic activity in in vivo and in vitro models of hepatic fibrosis. Although ISO is a direct metabolite of QCT differing by a methyl group, it has been reported to be absorbed more adequately and eliminated slower than QCT after oral administration. Our aim of the study was to investigate biological effect of mono-methylated QCT derivatives against fibrosis using rat hepatic stellate cells (HSC-T6). All test derivatives were synthesized from QCT. HSC-T6 cells were induced by TGFβ and treated with derivatives followed by cell proliferation assay, immunofluorescence staining of αSMA, and gene expression analysis of fibrosis markers. All compounds showed a dose- and time-dependent antiproliferation effect. ISO, 3-O-methylquercetin (3MQ), and rhamnetin (RHA) reduced αSMA mRNA; 3MQ prevented the augmentation of collagen I mRNA; and compounds, except azaleatin and 3MQ, reduced Timp1 mRNA expression in TGFβ-induced HSCs. In conclusion, each compound had singular effect against different features of fibrosis depending on the position of methyl group although the further mechanism of action of compounds during fibrosis development remains to be investigated. These findings suggest that antifibrotic effect of quercetin can be enhanced by adding methyl group on functionally important position.
Collapse
|
42
|
Hu J, Zhang Y, Jiang X, Zhang H, Gao Z, Li Y, Fu R, Li L, Li J, Cui H, Gao N. ROS-mediated activation and mitochondrial translocation of CaMKII contributes to Drp1-dependent mitochondrial fission and apoptosis in triple-negative breast cancer cells by isorhamnetin and chloroquine. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:225. [PMID: 31138329 PMCID: PMC6540563 DOI: 10.1186/s13046-019-1201-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022]
Abstract
Background Triple-negative breast cancer (TNBC) is often aggressive and associated with a poor prognosis. Due to the lack of available targeted therapies and to problems of resistance with conventional chemotherapeutic agents, finding new treatments for TNBC remains a challenge and a better therapeutic strategy is urgently required. Methods TNBC cells and xenograft mice were treated with a combination of chloroquine (CQ) and isorhamnetin (IH). Mitochondrial fission, apoptosis, and related signaling pathways were determined by flow cytometry, immunofluorescence, and related molecular biological techniques. Results The inhibition of autophagy/mitophagy by CQ selectively enhances IH-induced mitochondrial fission and apoptosis in TNBC cells but not in estrogen-dependent breast cancer cells. These events were accompanied by mitochondrial translocation of Bax and the release of cytochrome c. Mechanistically, these effects were associated with oxidative stress-mediated phosphorylation of CaMKII (Thr286) and Drp1 (S616), and subsequent mitochondrial translocation of CaMKII and Drp1. The interruption of the CaMKII pathway by genetic approaches (e.g. CaMKII mutant or siRNA) attenuated combination-mediated mitochondrial fission and apoptosis. The combination of CQ/IH was a marked inhibitor tumor growth, inducing apoptosis in the TNBC xenograft mouse model in association with the activation of CaMKII and Drp1 (S616). Conclusions Our study highlights the critical role of ROS-mediating CaMKII/Drp1 signaling in the regulation of mitochondrial fission and apoptosis induced by combination of CQ/IH. These findings also suggest that IH could potentially be further developed as a novel chemotherapeutic agent. Furthermore, a combination of IH with classic autophagy/mitophagy inhibitor could represent a novel therapeutic strategy for the treatment of TNBC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1201-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinjiao Hu
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yanhao Zhang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiuxing Jiang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Hongwei Zhang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Ziyi Gao
- Greater Philadelphia Pharmacy, Philadelphia, USA
| | - Yunong Li
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Ruoqiu Fu
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Lirong Li
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jie Li
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 2#Tiansheng Road, Beibei District, Chongqing, 400716, China. .,Medical Research Institute, Southwest University, 2#Tiansheng Road, Beibei District, Chongqing, 400716, China.
| | - Ning Gao
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
43
|
Wei S, Zhou X, Niu M, Zhang H, Liu X, Wang R, Li P, Li H, Cai H, Zhao Y. Network pharmacology exploration reveals the bioactive compounds and molecular mechanisms of Li-Ru-Kang against hyperplasia of mammary gland. Mol Genet Genomics 2019; 294:1159-1171. [PMID: 31053932 DOI: 10.1007/s00438-019-01569-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022]
Abstract
Li-Ru-Kang (LRK) has been commonly used in the treatment of hyperplasia of mammary gland (HMG) as a cipher prescription and achieved obvious therapeutic effects. However, the bioactive compounds and underlying pharmacological mechanisms remain unclear. This study aims to decipher the bioactive compounds and potential action mechanisms of LRK in the treatment of HMG using an integrated pharmacology approach. The ingredients of LRK and the corresponding drug targets were retrieved through drug target databases and were used to construct the "compound-target-disease" network and function-pathway network. Ultimately, 89 compounds and 2150 drug targets were collected. Gene ontology enrichment analysis revealed that mammary gland alveolus development and mammary gland lobule development were the key biological processes and were regulated simultaneously by three direct targets, including androgen receptor (AR), estrogen receptor (ER) and cyclin-D1. Moreover, 14 compounds of LRK were directly involved in the regulation of the three aforementioned targets. KEGG pathway enrichment analysis found that five signaling pathways and seven direct targets were closely related with HMG treatment by LRK. The results of animal experiments showed that LRK significantly improved the histopathological status of HMG in rats. Additionally, LRK markedly regulated the protein expressions of AR, cyclin-D1, MMP2, MMP3 and MMP9. But interestingly, the effect of LRK on ER was not obvious. This study demonstrated that LRK exerted its therapeutic efficacy based on multi-components, multi-targets and multi-pathways. This research confirms the advantages of network pharmacology analyses and the necessity for experimental verification.
Collapse
Affiliation(s)
- Shizhang Wei
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Xuelin Zhou
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Haizhu Zhang
- College of Pharmacy and Chemistry, Dali University, Dali, 671000, China
| | - Xiaoyi Liu
- China Military Institute of Chinese Medicine, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Ruilin Wang
- Department of Integrative Medical Center, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Pengyan Li
- China Military Institute of Chinese Medicine, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Haotian Li
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Huadan Cai
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Yanling Zhao
- Department of Pharmacy, the Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China.
| |
Collapse
|
44
|
Jiang H, Yamashita Y, Nakamura A, Croft K, Ashida H. Quercetin and its metabolite isorhamnetin promote glucose uptake through different signalling pathways in myotubes. Sci Rep 2019; 9:2690. [PMID: 30804434 PMCID: PMC6389993 DOI: 10.1038/s41598-019-38711-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022] Open
Abstract
Quercetin and its metabolite isorhamnetin elicit various beneficial effects on human health. However, their bioavailability is low. In this study, we investigated whether low concentrations in the physiological range could promote glucose uptake in L6 myotubes, as well as the underlying molecular mechanisms. We found that 0.1 nM and 1 nM quercetin or 1 nM isorhamnetin significantly increased glucose uptake via translocation of glucose transporter type 4 (GLUT4) to the plasma membrane of L6 myotubes. Quercetin principally activated the CaMKKβ/AMPK signalling pathway at these concentrations, but also activated IRS1/PI3K/Akt signalling at 10 nM. In contrast, 1 nM and 10 nM isorhamnetin principally activated the JAK/STAT pathway. Treatment with siAMPKα and siJAK2 abolished quercetin- and isorhamnetin-induced GLUT4 translocation, respectively. However, treatment with siJAK3 did not affect isorhamnetin-induced GLUT4 translocation, indicating that isorhamnetin induced GLUT4 translocation mainly through JAK2, but not JAK3, signalling. Thus, quercetin preferably activated the AMPK pathway and, accordingly, stimulated IRS1/PI3K/Akt signalling, while isorhamnetin activated the JAK2/STAT pathway. Furthermore, after oral administration of quercetin glycoside at 10 and 100 mg/kg body weight significantly induced GLUT4 translocation to the plasma membrane of skeletal muscles in mice. In the same animals, plasma concentrations of quercetin aglycone form were 4.95 and 6.80 nM, respectively. In conclusion, at low-concentration ranges, quercetin and isorhamnetin promote glucose uptake by increasing GLUT4 translocation via different signalling pathways in skeletal muscle cells; thus, these compounds may possess beneficial functions for maintaining glucose homeostasis by preventing hyperglycaemia at physiological concentrations.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Asuka Nakamura
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Kevin Croft
- School of Biomedical Science, The University of Western Australia, Perth, WA, 6009, Australia
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan.
| |
Collapse
|
45
|
Zhou P, Liu P, Zhang J. Long noncoding RNA RUSC1‑AS‑N promotes cell proliferation and metastasis through Wnt/β‑catenin signaling in human breast cancer. Mol Med Rep 2018; 19:861-868. [PMID: 30569097 PMCID: PMC6323231 DOI: 10.3892/mmr.2018.9763] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 10/03/2018] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is one of the most frequently diagnosed cancers among females worldwide. Long noncoding RNAs (lncRNAs) have been revealed to serve significant roles in diagnosis and treatment of breast cancer. In the present study, the novel lncRNA RUSC1-AS-N was demonstrated to promote cell viability and metastasis. A total of 100 patients with breast cancer were recruited for this study and it was revealed that RUSC1-AS-N was upregulated in tumor tissues compared with in adjacent non-cancerous counterparts. In addition, using several breast cancer cell lines, it was demonstrated that the mRNA levels of RUSC1-AS-N were highest in the notably metastatic cell lines MDA-MB-231 and MDA-MB-468. Knockdown of RUSC1-AS-N in breast cancer cells inhibited cell proliferation in the colony formation and cell proliferation assays. Furthermore, depletion of RUSC1-AS-N suppressed cell metastasis, as revealed by wound-healing and western blot assays. In addition, the protein levels of Wnt1 and β-catenin were significantly decreased when RUSC1-AS-N was knocked down. However, Wnt signaling pathway activator Wnt agonist 1 reversed the effects of RUSC1-AS-N knockdown on cell proliferation and metastasis. The present study demonstrated that lncRNA RUSC1-AS-N promoted cell viability and metastasis via Wnt/β-catenin signaling in human breast cancer, which may indicate novel targets for the treatment of breast cancer in clinic.
Collapse
Affiliation(s)
- Peng Zhou
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Peng Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Jin Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| |
Collapse
|
46
|
Exploring the Pharmacological Mechanism of Danzhi Xiaoyao Powder on ER-Positive Breast Cancer by a Network Pharmacology Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5059743. [PMID: 29692855 PMCID: PMC5859839 DOI: 10.1155/2018/5059743] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 01/16/2018] [Indexed: 12/14/2022]
Abstract
Background Breast cancer is the most common malignancy among women worldwide, but the long-term endocrine therapy is frequently associated with adverse side effects. Danzhi Xiaoyao powder (DXP) is a herbal formula that has an effect on breast cancer, especially ER-positive breast cancer. However, the active compounds, potential targets, and pharmacological and molecular mechanism of its action against cancer remain unclear. Methods A network pharmacology approach comprising drug-likeness evaluation, oral bioavailability prediction, Caco-2 permeability prediction, multiple compound target prediction, multiple known target collection, breast cancer genes collection, and network analysis has been used in this study. Results Four networks are set up—namely, ER-positive breast cancer network, compound-compound target network of DXP, DXP-ER-positive breast cancer network, and compound-known target-ER-positive breast cancer network. Some ER-positive breast cancer and DXP related targets, clusters, biological processes, and pathways, and several potential anticancer compounds are found. Conclusion This network analysis successfully predicted, illuminated, and confirmed the molecular synergy of DXP for ER-positive breast cancer, got potential anticancer active compounds, and found the potential ER-positive breast cancer associated targets, cluster, biological processes, and pathways. This work also provides clues to the researcher who explores ethnopharmacological or/and herbal medicine's or even multidrugs' various synergies.
Collapse
|
47
|
Meng LQ, Liu C, Luo YH, Piao XJ, Wang Y, Zhang Y, Wang JR, Wang H, Xu WT, Liu Y, Wu YQ, Sun HN, Han YH, Jin MH, Shen GN, Zang YQ, Li J, Fang NZ, Cui YD, Jin CH. Quinalizarin exerts an anti-tumour effect on lung cancer A549 cells by modulating the Akt, MAPK, STAT3 and p53 signalling pathways. Mol Med Rep 2017; 17:2626-2634. [PMID: 29207064 DOI: 10.3892/mmr.2017.8110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 11/09/2017] [Indexed: 11/06/2022] Open
Abstract
Quinalizarin may be a potential chemical agent for cancer therapy, as it exerts anti‑tumour effects against a variety of different types of cancer. However, the underlying regulatory mechanism and signalling pathways of quinalizarin in lung cancer cells remains unknown. The present study sought to investigate the effects of quinalizarin on proliferation, apoptosis and reactive oxygen species (ROS) generation in lung cancer. MTT assays were used to evaluate the effects of quinalizarin on the viability of lung cancer A549, NCI‑H460 and NCI‑H23 cells. Flow cytometry was employed to evaluate the effects of quinalizarin on the cell cycle, apoptosis and ROS generation in A549 cells. Western blotting was performed to detect cell cycle and apoptosis‑associated protein expression levels in A549 cells. Quinalizarin inhibited A549, NCI‑H460 and NCI‑H23 cell proliferation and induced A549 cell cycle arrest at the G0/G1 phase. Quinalizarin induced apoptosis by upregulating the expression of B‑cell lymphoma 2 (Bcl‑2)‑associated agonist of cell death, cleaved‑caspase‑3 and cleaved‑poly (adenosine diphosphate‑ribose) polymerase, and downregulating the expression of Bcl‑2. Furthermore, quinalizarin activated mitogen‑activated protein kinase (MAPK) and p53, and inhibited the protein kinase B and signal transducer and activator of transcription‑3 (STAT3) signalling pathways. In addition, quinalizarin increased ROS generation. The ROS scavenger N‑acetyl‑L‑cysteine restored quinalizarin‑induced cell apoptosis, and inactivated the MAPK and STAT3 signalling pathways. The results of the present study demonstrated that quinalizarin induces G0/G1 phase cell cycle arrest and apoptosis via ROS mediated‑MAPK and STAT3 signalling pathways.
Collapse
Affiliation(s)
- Ling-Qi Meng
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Chang Liu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hua Luo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Xian-Ji Piao
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang 163316, P.R. China
| | - Yue Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yang Liu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yi-Qin Wu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hu-Nan Sun
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hao Han
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Mei-Hua Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Gui-Nan Shen
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yan-Qing Zang
- College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jing Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Nan-Zhu Fang
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Yu-Dong Cui
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| |
Collapse
|
48
|
Park JY, Kim SI, Lee HJ, Kim SS, Kwon YS, Chun W. Isorhamnetin-3-O-Glucuronide Suppresses JNK and p38 Activation and Increases Heme-Oxygenase-1 in Lipopolysaccharide-Challenged RAW264.7 Cells. Drug Dev Res 2017; 77:143-51. [PMID: 27113811 DOI: 10.1002/ddr.21301] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/08/2016] [Indexed: 12/24/2022]
Abstract
Preclinical Research Isorhanmetin (ISH) exhibits a wide range of biological properties including anticancer, anti-oxidant and anti-inflammatory activities. However, the pharmacological properties of isorhamnetin-3-O-glucuronide (IG), a glycoside derivative of ISH, have not been extensively examined. The objective of this study was to examine the anti-inflammatory properties of IG and its underlying mechanism in lipopolysaccharide (LPS)-challenged RAW264.7 macrophage cells in comparison with its aglycone, ISH. IG suppressed LPS-induced extracellular secretion of the proinflammatory mediators, nitric oxide (NO) and PGE2 , and proinflammatory protein expressions of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2. IG also increased expression of heme oxygenase-1 (HO-1). IG attenuated LPS-induced activation of c-Jun N-terminal kinase (JNK) and p38 in a concentration-dependent manner with negligible suppression of extracellular signal-regulated kinases (ERK) phosphorylation. In conclusion, this study demonstrates that IG exerts anti-inflammatory activity by increasing HO-1 expression and by suppressing JNK and p38 signaling pathways in LPS-challenged RAW264.7 macrophage cells. Drug Dev Res 77 : 143-151, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jin-Young Park
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| | - Song-In Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| | - Sung-Soo Kim
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University, Chuncheon, 200-701, Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, 200-701, Korea
| |
Collapse
|
49
|
Gao L, Yao R, Liu Y, Wang Z, Huang Z, Du B, Zhang D, Wu L, Xiao L, Zhang Y. Isorhamnetin protects against cardiac hypertrophy through blocking PI3K-AKT pathway. Mol Cell Biochem 2017; 429:167-177. [PMID: 28176246 DOI: 10.1007/s11010-017-2944-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/17/2017] [Indexed: 01/10/2023]
Abstract
Isorhamnetin, a flavonoid compound extracted from the Chinese herb Hippophae rhamnoides L., is well known for its anti-inflammatory, anti-oxidative, anti-adipogenic, anti-proliferative, and anti-tumor activities. However, the role of isorhamnetin in cardiac hypertrophy has not been reported. The aims of the present study were to find whether isorhamnetin could alleviate cardiac hypertrophy and to define the underlying molecular mechanisms. Here, we investigated the effects of isorhamnetin (100 mg/kg/day) on cardiac hypertrophy induced by aortic banding in mice. Cardiac hypertrophy was evaluated by echocardiographic, hemodynamic, pathological, and molecular analyses. Our data demonstrated that isorhamnetin could inhibit cardiac hypertrophy and fibrosis 8 weeks after aortic banding. The results further revealed that the effect of isorhamnetin on cardiac hypertrophy was mediated by blocking the activation of phosphatidylinositol 3-kinase-AKT signaling pathway. In vitro studies performed in neonatal rat cardiomyocytes confirmed that isorhamnetin could attenuate cardiomyocyte hypertrophy induced by angiotensin II, which was associated with phosphatidylinositol 3-kinase-AKT signaling pathway. In conclusion, these data indicate for the first time that isorhamnetin has protective potential for targeting cardiac hypertrophy by blocking the phosphatidylinositol 3-kinase-AKT signaling pathway. Thus, our study suggests that isorhamnetin may represent a potential therapeutic strategy for the treatment of cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Rui Yao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Yuzhou Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Zheng Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Zhen Huang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Binbin Du
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Dianhong Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Leiming Wu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Lili Xiao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China
| | - Yanzhou Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, China.
| |
Collapse
|
50
|
Seo S, Seo K, Ki SH, Shin SM. Isorhamnetin Inhibits Reactive Oxygen Species-Dependent Hypoxia Inducible Factor (HIF)-1α Accumulation. Biol Pharm Bull 2017; 39:1830-1838. [PMID: 27803454 DOI: 10.1248/bpb.b16-00414] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Isorhamnetin is a flavonoid metabolite of quercetin and isolated from water dropwort (Oenanthe javanica, Umbelliferae). It has been reported that isorhamnetin exerts beneficial effects including antioxidant, anti-inflammatory, and anti-proliferative activities. The present study investigated whether the antioxidant activity of isorhamnetin is correlated with its anti-cancer effects on colorectal cancer cells. Isorhamnetin significantly repressed cobalt chloride (CoCl2)- or hypoxia-induced hypoxia inducible factor-1α (HIF-1α) accumulation in HCT116 and HT29 cells. When compared with quercetin, isorhamnetin showed potent inhibition of HIF-1α. Moreover, it inhibited CoCl2-induced activity of hypoxia response element reporter gene and HIF-1α-dependent transcription of genes such as glucose transporter 1, lactate dehydrogenase A, carbonic anhydrase-IX, and pyruvate dehydrogenase kinase 1. Isorhamnetin also blocked hydrogen peroxide (H2O2)-induced HIF-1α accumulation. The antioxidant effects of isorhamnetin were confirmed by observation of CoCl2- or H2O2-induced reactive oxygen species (ROS) production. Consistently, overexpressed HIF-1α was decreased by isorhamnetin or N-acetyl-L-cysteine in HEK293 cells. In vitro migration and invasion assay further confirmed the inhibitory effects of isorhamnetin on cancer cells. Collectively, these results demonstrate that isorhamnetin inhibits ROS-mediated HIF-1α accumulation, which contributes to its anti-metastatic efficacy.
Collapse
Affiliation(s)
- Suho Seo
- College of Pharmacy, Chosun University
| | | | | | | |
Collapse
|