1
|
Nguyen MH, Le NTH, Nguyen BQH, Nguyen MTT, Do TNV, Le TH, Nguyen VT, Yen CH. In vitro and in silico hybrid approach to unveil triterpenoids from Helicteres hirsuta leaves as potential compounds for inhibiting Nrf2. RSC Adv 2025; 15:1915-1923. [PMID: 39839230 PMCID: PMC11749225 DOI: 10.1039/d4ra07646j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/28/2024] [Indexed: 01/23/2025] Open
Abstract
Cancer is a leading global health concern, with over 20 million new cases and 9.7 million deaths reported in 2022. Chemotherapy remains a widely used treatment, but drug resistance, which affects up to 90% of treatment outcomes, significantly hampers its effectiveness. The transcription factor Nrf2, which is crucial for cellular defense against oxidative stress, plays a dual role in cancer treatment. Although Nrf2 activation can suppress early carcinogenesis, its overexpression in cancer cells contributes to drug resistance, resulting in poor patient outcomes. Thus, inhibiting Nrf2 has emerged as a promising strategy for overcoming cancer drug resistance. Natural compounds such as luteolin and brusatol have shown potential in inhibiting Nrf2, although with limitations. This study isolates and characterizes seven triterpenoids from the n-hexane sub-fraction of Helicteres hirsuta, a plant traditionally used for medicinal purposes, to evaluate their ability to modulate Nrf2 activity in Huh7 cancer and HaCaT normal cells. Additionally, molecular docking and dynamic simulations were utilized to assess the binding potential of these compounds to the PI3Kα receptor, which regulates downstream signaling pathways, thereby suppressing Nrf2 activity in cancer cells. Our findings provide insights into new strategies seeking triterpenoids as promising structures to reverse chemoresistance by regulating Nrf2. The results also reveal the potential of 3β-O-trans-caffeoylbetulinic acid from H. hirsuta leaves as the unprecedented compound inhibiting Nrf2 activity, with an IC50 of 74.5 μg mL-1 in Huh7 cancer cells.
Collapse
Affiliation(s)
- Minh Hien Nguyen
- University of Health Sciences, Vietnam National University Ho Chi Minh City YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward Di An City Binh Duong Province 75308 Vietnam (+84) 373 696 894
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
| | - Nguyen Thien Han Le
- University of Health Sciences, Vietnam National University Ho Chi Minh City YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward Di An City Binh Duong Province 75308 Vietnam (+84) 373 696 894
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
| | - Bui Quoc Huy Nguyen
- The University of Danang - VN-UK Institute for Research and Executive Education 41 Le Duan Street, Hai Chau 1 Ward, Hai Chau District Danang City 50000 Vietnam
| | - Mai Thanh Thi Nguyen
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
- Faculty of Chemistry, University of Science Ho Chi Minh City Vietnam
- Research Lab for Drug Discovery and Development, University of Science Ho Chi Minh City Vietnam
| | - Truong Nhat Van Do
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
- Faculty of Chemistry, University of Science Ho Chi Minh City Vietnam
- Research Lab for Drug Discovery and Development, University of Science Ho Chi Minh City Vietnam
| | - Tho Huu Le
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
- Faculty of Chemistry, University of Science Ho Chi Minh City Vietnam
- Research Lab for Drug Discovery and Development, University of Science Ho Chi Minh City Vietnam
| | - Vu Thanh Nguyen
- Division of Aquacultural Biotechnology, Biotechnology Center of Ho Chi Minh City 2374, Highway 1, Quarter 2, Trung My Tay Ward, District 12 Ho Chi Minh City 70000 Vietnam
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University No. 100, Shih-Chuan 1st Road, Sanmin District Kaohsiung City 80708 Taiwan
- National Natural Product Libraries and High-Throughput Screening Core Facility, Kaohsiung Medical University No. 100, Shih-Chuan 1st Road, Sanmin District Kaohsiung City 80708 Taiwan
| |
Collapse
|
2
|
Nguyen MH, Nguyen TYN, Le THN, Le TNT, Chau NTN, Le TMH, Huy Nguyen BQ. Medicinal plants as a potential resource for the discovery of novel structures towards cancer drug resistance treatment. Heliyon 2024; 10:e39229. [PMID: 39492898 PMCID: PMC11530815 DOI: 10.1016/j.heliyon.2024.e39229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/23/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Despite extensive research in chemotherapy, global cancer concerns persist, exacerbated by the challenge of drug resistance, which imposes economic and medical burdens. Natural compounds, particularly secondary metabolites from medicinal plants, present promising avenues for overcoming cancer drug resistance due to their diverse structures and essential pharmacological effects. This review provides a comprehensive exploration of cancer cell resistance mechanisms and target actions for reversing resistance and highlights the in vitro and in vivo efficacy of noteworthy alkaloids, flavonoids, and other compounds, emphasizing their potential as therapeutic agents. The molecular properties supporting ligand interactions are thoroughly examined, providing a robust theoretical foundation. The review concludes by discussing methods including quantitative structure-activity relationships and molecular docking, offering insights into screening potential candidates. Current trends in clinical treatment, contributing to a holistic understanding of the multifaceted approaches to address cancer drug resistance are also outlined.
Collapse
Affiliation(s)
- Minh Hien Nguyen
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
- Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc City, Ho Chi Minh city, Viet Nam
| | - Thi Yen Nhi Nguyen
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
- Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc City, Ho Chi Minh city, Viet Nam
- Faculty of Applied Science, Ho Chi Minh City University of Technology, Vietnam National University Ho Chi Minh City, 268 Ly Thuong Kiet Street Ward 14, District 10, Ho Chi Minh City, Viet Nam
| | - Thien Han Nguyen Le
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
| | - Thi Ngoc Tam Le
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
| | - Ngoc Trong Nghia Chau
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
| | - Tu Manh Huy Le
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
| | - Bui Quoc Huy Nguyen
- The University of Danang - VN-UK Institute for Research and Executive Education, 41 Le Duan Street, Hai Chau 1 Ward, Hai Chau District, Danang City, Viet Nam
| |
Collapse
|
3
|
Das A, Biggs MA, Hunt HL, Mahabadi V, Goncalves BG, Phan CAN, Banerjee IA. Design and investigation of novel iridoid-based peptide conjugates for targeting EGFR and its mutants L858R and T790M/L858R/C797S: an in silico study. Mol Divers 2024:10.1007/s11030-024-11007-3. [PMID: 39424745 DOI: 10.1007/s11030-024-11007-3] [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: 06/09/2024] [Accepted: 09/29/2024] [Indexed: 10/21/2024]
Abstract
In this work, we designed novel peptide conjugates with plant-based iridoid and lichen-derived depside derivatives to target the wild-type EGFR (WT) and its mutants, L858R and T790M/L858R/C797S triple mutant. These mutations are often expressed in multiple cancers, particularly lung cancer. Specifically, the iridoids included 7-deoxyloganetic acid (7-DGA) and loganic acid (LG), while the depside derivative was sekikaic acid (SK). These compounds are known for their innate anticancer properties and were conjugated with two separate peptide sequences KLPGWSG (K) and YSIPKSS (Y). These sequences have been shown to target EGFR in previous phage display library screening, although the mechanism is unknown. Thus, we created the di-conjugates for dual targeting and investigated their interactions of the di-conjugates and that of the neat peptides with the kinase domain of EGFR (WT) and the two mutants using molecular docking, molecular dynamics (MD) simulations, and MM-GBSA analysis. Docking studies revealed that the (7-DGA)2-K showed the highest binding affinity at - 9.3 kcal/mol with the L858R mutant, while (LG)2-Y displayed the highest binding affinity at - 9.0 kcal/mol for the triple mutant receptor. Our results indicated that several of the conjugates interacted with crucial residues of the kinase domain, including ASP855 and THR854 (activation loop), MET793 and PRO794 (hinge region), ARG841 (catalytic loop), and LYS728 and LEU718 of the glycine-rich P-loop. Interestingly, strong hydrophobic interactions were also observed with the C-terminal tail residues, such as PHE997 and ALA1000 as well as with ARG999 for the YSIPKSS peptide and most of the conjugates. The hydroxyl group of the cyclopentane ring and the oxygen of the pyran ring of the (7-DGA)2-peptide conjugates contributed to binding particularly in the hinge region, while the peptide components formed an extended structure that bound well into the C-lobe. The (SK)2-Y di-conjugate and KLPGWSG peptide formed hydrogen bonds with the SER797 residue of the triple mutant. Overall, our results show that the (7-DGA)2-K, di-conjugate, the (7-DGA)2-Y di-conjugate, and the neat YSIPKSS demonstrated strong and stable binding with the L858R mutant and the highly resistant triple mutant EGFR, respectively. The novel designed conjugates demonstrate potential for further optimization for laboratory studies aimed at developing new therapeutics for targeting specific EGFR mutant expressing cells.
Collapse
Affiliation(s)
- Amrita Das
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Mary A Biggs
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Hannah L Hunt
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Vida Mahabadi
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Beatriz G Goncalves
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Chau Anh N Phan
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
4
|
Ma J, Mo J, Feng Y, Wang L, Jiang H, Li J, Jin C. Combination of transcriptomic and proteomic approaches helps unravel the mechanisms of luteolin in inducing liver cancer cell death via targeting AKT1 and SRC. Front Pharmacol 2024; 15:1450847. [PMID: 39234106 PMCID: PMC11371790 DOI: 10.3389/fphar.2024.1450847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Luteolin, a natural compound commonly used in traditional Chinese medicine, shows clinical potential as an anti-liver cancer agent. The mechanisms underlying the anti-liver cancer effect of luteolin are limited versus those reported for other cancers. Accordingly, this study was conducted to bridge the existing knowledge gap. Methods Transcriptomic and proteomic analyses of the response of the hepatocellular carcinoma cell line HuH-7 to luteolin were conducted, and a possible pathway was elucidated using confocal laser scanning microscopy (CLSM), flow cytometry, western blotting, qRT-PCR and bio-layer interferometry assay to systematically explore the possible mechanisms underlying the inhibition of the proliferation of liver cancer cells by luteolin. Results and Discussion Results showed that luteolin significantly inhibited HuH-7 cell proliferation. Transcriptomic and proteomic analyses collectively revealed that luteolin could promote cell cycle arrest and apoptosis in HuH-7 cells through transcription factors p53, nuclear factor kappa B (NF-κB), FOXO, ATF2, and TCF/LEF via AKT1, as well as the KEAP-NRF and SRC-STAT3 pathways. Furthermore, AKT1 and SRC were identified as the 2 targets of luteolin. Nuclear translocation of transcription factors p53 and NF-κB were affected by luteolin administration. Additionally, AKT1 activity affected normal metabolism in HuH-7 cells and resulted in the accumulation of reactive oxygen species, which activated MOMP and further promoted apoptosis. Our results systematically elucidate the mechanism of luteolin in inhibiting the proliferation of liver cancer cells, mainly through cell cycle arrest and apoptosis via targeting AKT1 and SRC.
Collapse
Affiliation(s)
- Junxia Ma
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Evolutionary Ecology and Conservation, Taizhou University, Taizhou, China
| | - Jinggang Mo
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Yifu Feng
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Liezhi Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Hao Jiang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Junmin Li
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Evolutionary Ecology and Conservation, Taizhou University, Taizhou, China
| | - Chong Jin
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
5
|
Zhang J, Ma Y. Luteolin as a potential therapeutic candidate for lung cancer: Emerging preclinical evidence. Biomed Pharmacother 2024; 176:116909. [PMID: 38852513 DOI: 10.1016/j.biopha.2024.116909] [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: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Lung cancer is a prevalent malignant tumor and a leading cause of cancer-related fatalities globally. However, current treatments all have limitations. Therefore, there is an urgent need to identify a readily available therapeutic agent to counteract lung cancer development and progression. Luteolin is a flavonoid derived from vegetables and herbs that possesses preventive and therapeutic effects on various cancers. With the goal of providing new directions for the treatment of lung cancer, we review here the recent findings on luteolin so as to provide new ideas for the development of new anti-lung cancer drugs. The search focused on studies published between January 1995 and January 2024 that explored the use of luteolin in lung cancer. A comprehensive literature search was conducted in the SCOPUS, Google Scholar, PubMed, and Web of Science databases using the keywords "luteolin" and "lung cancer." By collecting previous literature, we found that luteolin has multiple mechanisms of therapeutic effects, including promotion of apoptosis in lung cancer cells; inhibition of tumor cell proliferation, invasion and metastasis; and modulation of immune responses. In addition, it can be used as an adjuvant to radio-chemotherapy and helps to ameliorate cancer complications. This review summarizes the structure, natural sources, physicochemical properties and pharmacokinetics of luteolin, and focuses on the anti-lung cancer mechanism of luteolin, so as to provide new ideas for the development of new anti-lung cancer drugs.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China
| | - Yue Ma
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China.
| |
Collapse
|
6
|
Emran TB, Eva TA, Zehravi M, Islam F, Khan J, Kareemulla S, Arjun UVNV, Balakrishnan A, Taru PP, Nainu F, Salim E, Rab SO, Nafady MH, Wilairatana P, Park MN, Kim B. Polyphenols as Therapeutics in Respiratory Diseases: Moving from Preclinical Evidence to Potential Clinical Applications. Int J Biol Sci 2024; 20:3236-3256. [PMID: 38904027 PMCID: PMC11186353 DOI: 10.7150/ijbs.93875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/04/2024] [Indexed: 06/22/2024] Open
Abstract
Respiratory diseases are the most common and severe health complication and a leading cause of death worldwide. Despite breakthroughs in diagnosis and treatment, few safe and effective therapeutics have been reported. Phytochemicals are gaining popularity due to their beneficial effects and low toxicity. Polyphenols are secondary metabolites with high molecular weights found at high levels in natural food sources such as fruits, vegetables, grains, and citrus seeds. Over recent decades, polyphenols and their beneficial effects on human health have been the subject of intense research, with notable successes in preventing major chronic non-communicable diseases. Many respiratory syndromes can be treated effectively with polyphenolic supplements, including acute lung damage, pulmonary fibrosis, asthma, pulmonary hypertension, and lung cancer. This review summarizes the role of polyphenols in respiratory conditions with sufficient experimental data, highlights polyphenols with beneficial effects for each, and identifies those with therapeutic potential and their underlying mechanisms. Moreover, clinical studies and future research opportunities in this area are discussed.
Collapse
Affiliation(s)
- Talha Bin Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Taslima Akter Eva
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Jishan Khan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Shaik Kareemulla
- Department of Pharmacy Practice, M. M. College of Pharmacy (Maharishi Markandeshwar Deemed University), Mullana-Ambala, Haryana 133207, India
| | - Uppuluri Varuna Naga Venkata Arjun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology, and Advanced Studies (VISTAS), Tamil Nadu, India
| | - Anitha Balakrishnan
- Department of Pharmaceutics, GRT Institute of Pharmaceutical Education and Research, Tiruttani, India
| | - Poonam Popatrao Taru
- Department of Pharmacognosy, School of Pharmacy, Vishwakarma University, Kondhwa, Pune, India
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia
| | - Emil Salim
- Department of Pharmacology and Clinical/Community Pharmacy, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, 20155, Indonesia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohamed H. Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza 12568, Egypt
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02453, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02453, Republic of Korea
| |
Collapse
|
7
|
Lee K, Choi YJ, Lim HI, Cho KJ, Kang N, Ko SG. Network pharmacology study to explore the multiple molecular mechanism of SH003 in the treatment of non-small cell lung cancer. BMC Complement Med Ther 2024; 24:70. [PMID: 38303001 PMCID: PMC10832243 DOI: 10.1186/s12906-024-04347-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is one of the leading causes of human death worldwide. Herbal prescription SH003 has been developed to treat several cancers including NSCLC. Due to the multi-component nature of SH003 with multiple targets and pathways, a network pharmacology study was conducted to analyze its active compounds, potential targets, and pathways for the treatment of NSCLC. METHODS We systematically identified oral active compounds within SH003, employing ADME criteria-based screening from TM-MC, OASIS, and TCMSP databases. Concurrently, SH003-related and NSCLC-associated targets were amalgamated from various databases. Overlapping targets were deemed anti-NSCLC entities of SH003. Protein-protein interaction networks were constructed using the STRING database, allowing the identification of pivotal proteins through node centrality measures. Empirical validation was pursued through LC-MS analysis of active compounds. Additionally, in vitro experiments, such as MTT cell viability assays and western blot analyses, were conducted to corroborate network pharmacology findings. RESULTS We discerned 20 oral active compounds within SH003 and identified 239 core targets shared between SH003 and NSCLC-related genes. Network analyses spotlighted 79 hub genes, including TP53, JUN, AKT1, STAT3, and MAPK3, crucial in NSCLC treatment. GO and KEGG analyses underscored SH003's multifaceted anti-NSCLC effects from a genetic perspective. Experimental validations verified SH003's impact on NSCLC cell viability and the downregulation of hub genes. LC-MS analysis confirmed the presence of four active compounds, namely hispidulin, luteolin, baicalein, and chrysoeriol, among the eight compounds with a median of > 10 degrees in the herb-compounds-targets network in SH003. Previously unidentified targets like CASP9, MAPK9, and MCL1 were unveiled, supported by existing NSCLC literature, enhancing the pivotal role of empirical validation in network pharmacology. CONCLUSION Our study pioneers the harmonization of theoretical predictions with practical validations. Empirical validation illuminates specific SH003 compounds within NSCLC, simultaneously uncovering novel targets for NSCLC treatment. This integrated strategy, accentuating empirical validation, establishes a paradigm for in-depth herbal medicine exploration. Furthermore, our network pharmacology study unveils fresh insights into SH003's multifaceted molecular mechanisms combating NSCLC. Through this approach, we delineate active compounds of SH003 and target pathways, reshaping our understanding of its therapeutic mechanisms in NSCLC treatment.
Collapse
Affiliation(s)
- Kangwook Lee
- Department of Food and Biotechnology, Korea University, Sejong, 30019, South Korea
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Yu-Jeong Choi
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Hae-In Lim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Kwang Jin Cho
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Nuri Kang
- Department of Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea.
| |
Collapse
|
8
|
Adedokun KA, Imodoye SO, Yahaya ZS, Oyeyemi IT, Bello IO, Adeyemo‐Imodoye MT, Sanusi MA, Kamorudeen RT. Nanodelivery of Polyphenols as Nutraceuticals in Anticancer Interventions. POLYPHENOLS 2023:188-224. [DOI: 10.1002/9781394188864.ch10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Li X, Tang Y, Liang P, Sun M, Li T, Shen Z, Sha S. Luteolin inhibits A549 cells proliferation and migration by down-regulating androgen receptors. Eur J Med Res 2023; 28:353. [PMID: 37716981 PMCID: PMC10504720 DOI: 10.1186/s40001-023-01302-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/18/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Yi Fei Qing Hua Granules (YQG) is a traditional Chinese herbal medicine with the effects of inhibiting the proliferation of lung cancer cells. Luteolin is one of the active compounds of YQG. Luteolin is a common flavonoid extracted from natural herbs and it can promote cancer cells apoptosis has been reported. However, the underlying molecular mechanism and effects of luteolin on human lung cancer needs to be validated. METHODS Molecular docking, network pharmacology methods and quantitative structure-activity relationship (QSAR) model were used to identify the active components of YQG and their possible mechanisms of action. Western blot analysis was used to measure AR expression in A549 cells. Cell migration assays were used to detect A549 cells proliferation transfected by AR plasmid and AR mutation plasmid, respectively. RESULTS TCMSP search results revealed that there are 182 active compounds in YQG, which correspond to 232 target genes. Sixty-one genes were overlapping genes in the 2 datasets of TCMSP and GeneCards. Through bioinformatics tagging of these overlapping genes, a total of 1,951 GO functional tagging analysis and 133 KEGG pathways were obtained. Through molecular docking technology and QSAR model verification, the multi-target active compound luteolin was screened out as one of the active components of YQG for in vitro verification. Androgen receptor (AR) was the hub protein with the highest docking score of luteolin. Western blot showed that luteolin could inhibit AR protein expression in lung cancer cell line A549. After the phosphorylation site of AR protein 877 was inactivated, the ability of luteolin to inhibit the proliferation of lung cancer cells was weakened. Luteolin significantly inhibited the growth of A549 xenogeneic tumors at day 25 and 28 and inhibited the expression of AR. CONCLUSION In this study, we have explored luteolin as one of the active components of YQG, and may inhibit the proliferation and migration of A549 cells by decreasing the expression of AR and the regulation of phosphorylation at AR-binding sites.
Collapse
Affiliation(s)
- Xu Li
- Tongji University School of Medicine, Shanghai, 200092, China
- General practice, Tongji University School of Medicine Affiliated Anting Community Health Center of Jiading District, Shanghai, 201805, China
| | - Yeling Tang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Pengchen Liang
- School of Microelectronics, Shanghai University, Shanghai, 201800, China
| | - Miaomiao Sun
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Tian Li
- Graduate School, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhiping Shen
- General practice, Tongji University School of Medicine Affiliated Anting Community Health Center of Jiading District, Shanghai, 201805, China.
| | - Shuang Sha
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
10
|
Prasher P, Sharma M, Singh SK, Gulati M, Chellappan DK, Zacconi F, De Rubis G, Gupta G, Sharifi-Rad J, Cho WC, Dua K. Luteolin: a flavonoid with a multifaceted anticancer potential. Cancer Cell Int 2022; 22:386. [PMID: 36482329 PMCID: PMC9730645 DOI: 10.1186/s12935-022-02808-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Therapeutic effect of phytochemicals has been emphasized in the traditional medicine owing to the presence of bioactive molecules, such as polyphenols. Luteolin is a flavone belonging to the flavonoid class of polyphenolic phytochemicals with healing effect on hypertension, inflammatory disorders, and cancer due to its action as pro-oxidants and antioxidants. The anticancer profile of luteolin is of interest due to the toxic effect of contemporary chemotherapy paradigm, leading to the pressing need for the development and identification of physiologically benevolent anticancer agents and molecules. Luteolin exerts anticancer activity by downregulation of key regulatory pathways associated with oncogenesis, in addition to the induction of oxidative stress, cell cycle arrest, upregulation of apoptotic genes, and inhibition of cell proliferation and angiogenesis in cancer cells. In this review, we discuss about the anticancer profile of luteolin.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun, 248007 India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, 248007 India
| | - Sachin Kumar Singh
- School of Pharmacy and Pharmaceutical Science, Lovely Professional University, Phagwara, India
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
| | - Monica Gulati
- School of Pharmacy and Pharmaceutical Science, Lovely Professional University, Phagwara, India
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Flavia Zacconi
- Departamento de Quimica Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuna Mackenna 4860, Macul, 7820436 Santiago, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, 7820436 Santiago, Chile
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong China
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| |
Collapse
|
11
|
Flavones: Six Selected Flavones and Their Related Signaling Pathways That Induce Apoptosis in Cancer. Int J Mol Sci 2022; 23:ijms231810965. [PMID: 36142874 PMCID: PMC9505532 DOI: 10.3390/ijms231810965] [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: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer is a horrific disease that, to date, has no cure. It is caused by various factors and takes many lives. Apoptosis is a programmed cell death mechanism and if it does not function correctly in cancer cells, it can lead to severe disease. There are various signaling pathways for regulating apoptosis in cancer cells. Flavonoids are non-artificial natural bioactive compounds that are gaining attention as being capable of for inducing apoptosis in cancer cells. Among these, in this study, we focus on flavones. Flavones are a subclass of the numerous available flavonoids and possess several bioactive functions. Some of the most reported and well-known critical flavones, namely apigenin, acacetin, baicalein, luteolin, tangeretin, and wogonin, are discussed in depth in this review. Our main aim is to investigate the effects of the selected flavones on apoptosis and cell signaling pathways that contribute to death due to various types of cancers.
Collapse
|
12
|
Diedrich C, Camargo Zittlau I, Schineider Machado C, Taise Fin M, Maissar Khalil N, Badea I, Mara Mainardes R. Mucoadhesive nanoemulsion enhances brain bioavailability of luteolin after intranasal administration and induces apoptosis to sh-sy5y neuroblastoma cells. Int J Pharm 2022; 626:122142. [PMID: 36064075 DOI: 10.1016/j.ijpharm.2022.122142] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
Abstract
Neuroblastoma is the most frequently diagnosed extracranial solid tumor in children and accounts for 7% of all childhood malignancies and 15% cancer mortality in children. Luteolin (LUT) is recognized by its anticancer activity against several types of cancer. The aim of this study was to prepare chitosan-coated nanoemulsion containing luteolin (NECh-LUT), investigate its potential for brain delivery following intranasal administration, and to evaluate its cytotoxicity against neuroblastoma cells. NECh-LUT was developed by cavitation process and characterized for its size, surface charge, encapsulation efficiency, and mucoadhesion. The developed formulation presented size 68±1 nm, zeta potential +13±1 mV, and encapsulation efficiency of 85.5±0.3%. The NECh-LUT presented nearly 6-fold higher permeation through the nasal mucosa ex vivo and prolonged LUT release up to 72 h in vitro, following Baker-Lonsdale kinetic model. The pharmacokinetic evaluation of NECh-LUT revealed a 10-fold increase in drug half-life and a 4.4 times enhancement in LUT biodistribution in brain tissue after intranasal administration of single-dose. In addition, NECh-LUT inhibited the growth of neuroblastoma cells after 24, 48 and 72 h in concentrations starting from 2 µM. The NECh-LUT developed for intranasal administration proved to be a promising alternative for brain delivery of LUT, and a viable option for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Camila Diedrich
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Isabella Camargo Zittlau
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Christiane Schineider Machado
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Margani Taise Fin
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Najeh Maissar Khalil
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil
| | - Ildiko Badea
- Drug Design and Discovery Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Rubiana Mara Mainardes
- Pharmaceutical Nanotechnology Laboratory, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838 - CEP 85040-167, Guarapuava, PR, Brazil.
| |
Collapse
|
13
|
Pan J, Yang H, Zhu L, Lou Y, Jin B. Qingfei Jiedu decoction inhibits PD-L1 expression in lung adenocarcinoma based on network pharmacology analysis, molecular docking and experimental verification. Front Pharmacol 2022; 13:897966. [PMID: 36091822 PMCID: PMC9454399 DOI: 10.3389/fphar.2022.897966] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
Objective: We aim at investigating the molecular mechanisms through which the Qingfei Jiedu decoction (QFJDD) regulates PD-L1 expression in lung adenocarcinoma (LUAD). Methods: Bioactive compounds and targets of QFJDD were screened from TCMSP, BATMAN-TCM, and literature. Then, GeneCard, OMIM, PharmGKB, Therapeutic Target, and DrugBank databases were used to identify LUAD-related genes. The protein-protein interaction (PPI) network was constructed using overlapping targets of bioactive compounds in LUAD with the Cytoscape software and STRING database. The potential functions and pathways in which the hub genes were enriched by GO, KEGG, and DAVID pathway analyses. Molecular docking of bioactive compounds and key genes was executed via AutoDock Vina. Qualitative and quantitative analyses of QFJDD were performed using UPLC-Q-TOF-MS and UPLC. Expressions of key genes were determined by qRT-PCR, immunoreactivity score (IRS) of PD-L1 was assessed by immunohistochemistry (IHC), while the CD8+PD-1+T% derived from spleen tissues of Lewis lung cancer (LLC) bearing-mice was calculated using flow cytometry (FCM). Results: A total of 53 bioactive compounds and 288 targets of QFJDD as well as 8151 LUAD associated genes were obtained. Further, six bioactive compounds, including quercetin, luteolin, kaempferol, wogonin, baicalein, and acacetin, and 22 hub genes were identified. The GO analysis showed that the hub genes were mainly enriched in DNA or RNA transcription. KEGG and DAVID pathway analyses revealed that 20 hub genes were primarily enriched in virus, cancer, immune, endocrine, and cardiovascular pathways. The EGFR, JUN, RELA, HIF1A, NFKBIA, AKT1, MAPK1, and MAPK14 hub genes were identified as key genes in PD-L1 expression and PD-1 checkpoint pathway. Moreover, ideal affinity and regions were identified between core compounds and key genes. Notably, QFJDD downregulated EGFR, JUN, RELA, HIF1A, NFKBIA, and CD274 expressions (p < 0.05), while it upregulated AKT1 and MAPK1 (p < 0.05) levels in A549 cells. The PD-L1 IRS of LLC tissue in the QFJDD high dose (Hd) group was lower than model group (p < 0.01). CD8+PD-1+T% was higher in the QFJDD Hd group than in normal and model groups (p < 0.05). Conclusion: QFJDD downregulates PD-L1 expression and increases CD8+PD-1+T% via regulating HIF-1, EGFR, JUN and NFκB signaling pathways. Therefore, QFJDD is a potential treatment option for LUAD.
Collapse
Affiliation(s)
- Junjie Pan
- Department of Pulmonary and Critical Care Medicine, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Hangzhou, Zhejiang, China
- Department of Pulmonary and Critical Care Medicine, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Hongkuan Yang
- Respiratory Intensive Care Unit, The People’s Hospital of Gaozhou, Maoming, Guangdong, China
| | - Lihong Zhu
- Department of Pulmonary and Critical Care Medicine, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Hangzhou, Zhejiang, China
- Department of Pulmonary and Critical Care Medicine, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Yafang Lou
- Department of Pulmonary and Critical Care Medicine, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Hangzhou, Zhejiang, China
- Department of Pulmonary and Critical Care Medicine, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
- *Correspondence: Yafang Lou, ; Bo Jin,
| | - Bo Jin
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- *Correspondence: Yafang Lou, ; Bo Jin,
| |
Collapse
|
14
|
Yin FT, Zhou XH, Kang SY, Li XH, Li J, Ullah I, Zhang AH, Sun H, Wang XJ. Prediction of the mechanism of Dachengqi Decoction treating colorectal cancer based on the analysis method of " into serum components -action target-key pathway". JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115286. [PMID: 35413412 DOI: 10.1016/j.jep.2022.115286] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/21/2022] [Accepted: 04/06/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Colorectal cancer (CRC) is a common digestive tract malignant tumor that its morbidity and mortality seriously affect human health. At present, Dachengqi Decoction (DCQ), a traditional Chinese medicine formula, has been clinically used as an adjuvant therapy for CRC. However, pharmacodynamic substance basis and therapeutic mechanism are still unclear. AIM OF THE STUDY The main constituents absorbed in the blood and possible active targets after DCQ administration were explored based on the analysis method of "into serum components, action target and key pathway", which may provide reference for the study of the pharmacodynamic material basis and action mechanism of Dachengqi Decoction in the treatment of CRC. MATERIAL AND METHODS Based on the serum pharmacochemistry of traditional Chinese medicine (TCM), the prescription prototype ingredients of DCQ in mice serum samples were identified by ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry technology (UPLC-Q-TOF-MSE). Taking the prototype ingredients absorbed into serum as the research object, the possible targets and key pathways of DCQ in vivo were demonstrated by network pharmacology. Finally, using molecular docking verified the binding activity of prototype components and potential action targets. RESULTS A total of 46 prototype components of DCQ were identified in mice serum, most of which were derived from flavonoids and anthraquinones in Citrus aurantium L. and Rheum palmatum L. Network pharmacology prediction results indicated that the drug prototype components entering the serum may mainly regulate targets including mitogen-activated protein kinase (MAPK), interleukin-6 (IL-6), vascular endothelial growth factor (VEGF), etc. and main pathways such as (phosphatidylinositol 3-kinase/protein kinase B) PI3K-AKT signaling pathway, advanced glycation end products-receptor for AGE (AGE-RAGE) signaling pathway and IL-17 signaling pathway, etc. Molecular docking showed that the prototype active components had strong binding activity to VEGF, Harvey rat sarcoma viral oncogene homolog (HRAS) and MAPK1. CONCLUSIONS This study elucidated that most of the direct acting substances of DCQ in vivo were flavonoids and anthraquinones, which may play a role in regulating cell reproduction and apoptosis and inhibiting inflammation, providing a reference for the research of pharmacodynamic material basis and mechanism of DCQ in the treatment of CRC.
Collapse
Affiliation(s)
- Feng-Ting Yin
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Xiao-Hang Zhou
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Shu-Yu Kang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xing-Hua Li
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jing Li
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Ihsan Ullah
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Ai-Hua Zhang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Hui Sun
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xi-Jun Wang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
15
|
Hazafa A, Iqbal MO, Javaid U, Tareen MBK, Amna D, Ramzan A, Piracha S, Naeem M. Inhibitory effect of polyphenols (phenolic acids, lignans, and stilbenes) on cancer by regulating signal transduction pathways: a review. Clin Transl Oncol 2022; 24:432-445. [PMID: 34609675 DOI: 10.1007/s12094-021-02709-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/11/2021] [Indexed: 02/07/2023]
Abstract
Natural products, especially polyphenols (phenolic acids, lignans, and stilbenes) are suggested to be more potent anticancer drugs because of their no or less adverse effects, excess availability, high accuracy, and secure mode of action. In the present review, potential anticancer mechanisms of action of some polyphenols including phenolic acids, lignans, and stilbenes are discussed based on clinical, epidemiological, in vivo, and in vitro studies. The emerging evidence revealed that phenolic acids, lignans, and stilbenes induced apoptosis in the treatment of breast (MCF-7), colon (Caco-2), lung (SKLU-1), prostate (DU-145 and LNCaP), hepatocellular (hepG-2), and cervical (A-431) cancer cells, cell cycle arrest (S/G2/M/G1-phases) in gastric (MKN-45 and MKN-74), colorectal (HCT-116), bladder (T-24 and 5637), oral (H-400), leukemic (HL-60 and MOLT-4) and colon (Caco-2) cancer cells, and inhibit cell proliferation against the prostate (PC-3), liver (LI-90), breast (T47D and MDA-MB-231), colon (HT-29 and Caco-2), cervical (HTB-35), and MIC-1 cancer cells through caspase-3, MAPK, AMPK, Akt, NF-κB, Wnt, CD95, and SIRT1 pathways. Based on accumulated data, we suggested that polyphenols could be considered as a viable therapeutic option in the treatment of cancer cells in the near future.
Collapse
Affiliation(s)
- A Hazafa
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, 38040, Pakistan.
| | - M O Iqbal
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - U Javaid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - M B K Tareen
- College of Food Science & Technology, Huazhong Agricultural University, Huazhong, China
| | - D Amna
- Institute of Food Science & Nutrition, Bahauddin Zakariya University, Multan, Pakistan
| | - A Ramzan
- Department of Botany, University of Agriculture Faisalabad, Faisalabad, 38040, Pakistan
| | - S Piracha
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad, 38040, Pakistan
| | - M Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang, China
| |
Collapse
|
16
|
Zhang D, Zhang T, Zhang Y, Li Z, Li H, Zhang Y, Liu C, Han Z, Li J, Zhu J. Screening the components of Saussurea involucrata for novel targets for the treatment of NSCLC using network pharmacology. BMC Complement Med Ther 2022; 22:53. [PMID: 35227278 PMCID: PMC8886885 DOI: 10.1186/s12906-021-03501-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Saussurea involucrata (SAIN), also known as Snow lotus (SI), is mainly distributed in high-altitude areas such as Tibet and Xinjiang in China. To identify novel targets for the prevention or treatment of lung adenocarcinoma and lung squamous cell carcinoma (LUAD&LUSC), and to facilitate better alternative new drug discovery as well as clinical application services, the therapeutic effects of SAIN on LUAD&LUSC were evaluated by gene differential analysis of clinical samples, compound target molecular docking, and GROMACS molecular dynamics simulation. RESULTS Through data screening, alignment, analysis, and validation it was confirmed that three of the major active ingredients in SAIN, namely quercetin (Q), luteolin (L), and kaempferol (K), mainly act on six protein targets, which mainly regulate signaling pathways in cancer, transcriptional misregulation in cancer, EGFR tyrosine kinase inhibitor resistance, adherens junction, IL-17 signaling pathway, melanoma, and non-small cell lung cancer. In addition, microRNAs in cancer exert preventive or therapeutic effects on LUAD&LUSC. Molecular dynamics (MD) simulations of Q, L, or K in complex with EGFR, MET, MMP1, or MMP3 revealed the presence of Q in a very stable tertiary structure in the human body. CONCLUSION There are three active compounds of Q, L, and K in SAIN, which play a role in the treatment and prevention of non-small cell lung cancer (NSCLC) by directly or indirectly regulating the expression of genes such as MMP1, MMP3, and EGFR.
Collapse
Affiliation(s)
- Dongdong Zhang
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China
| | - Tieying Zhang
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China
| | - Yao Zhang
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China
| | - Zhongqing Li
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China
| | - He Li
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China
| | - Yueyang Zhang
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China
| | - Chenggong Liu
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China
| | - Zichao Han
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China
| | - Jin Li
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China.
| | - Jianbo Zhu
- School of Life Sciences, Shihezi University, Xiangyang street, Shihezi, 832003, PR China.
| |
Collapse
|
17
|
Tang X, Cheng L, Li G, Yan YM, Su F, Huang DL, Zhang S, Liu Z, Qian M, Li J, Cheng YX, Liu B. A small-molecule compound D6 overcomes EGFR-T790M-mediated resistance in non-small cell lung cancer. Commun Biol 2021; 4:1391. [PMID: 34903832 PMCID: PMC8668973 DOI: 10.1038/s42003-021-02906-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 11/16/2021] [Indexed: 11/10/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a deadly and highly prevalent malignancy. Targeting activated-EGFR mutations in NSCLC via EGFR tyrosine kinase inhibitor (EGFR-TKI) initially achieves a profound therapeutic response, but resistance frequently evolves, reducing treatment options. Here, we present a small-molecule compound D6 which selectively inhibits tumor cell growth and migration in NSCLC cells with EGFR-TKI-resistant T790M-EGFR-activated mutations (T790M-EGFR-AM), e.g., L858R/T790M, 19Del/T790M and L858R/T790M/C797S. D6 mimics a natural product isolated from the roots of Codonopsis pilosula and selectively competes with T790M-EGFR-AM to bind to HSP90, thus facilitating the ubiquitination dependent proteasomal degradation of T790M-EGFR-AM. By contrast, D6 has little impact on typical HSP90 chaperone activity, suggesting low systemic toxicity. Promisingly, D6 combined with erlotinib or osimertinib shows efficacy in overcoming the EGFR-TKIs-resistance in NSCLCs. Our study raises an alternative strategy to overcome T790M-mediated EGFR-TKI resistance in NSCLC via targeting the protein-protein interaction of HSP90 and T790M-EGFR by intervention with D6.
Collapse
Affiliation(s)
- Xiaolong Tang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China.
| | - Lizhi Cheng
- grid.263488.30000 0001 0472 9649Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Guo Li
- grid.452223.00000 0004 1757 7615Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Ming Yan
- grid.263488.30000 0001 0472 9649Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Fengting Su
- grid.263488.30000 0001 0472 9649Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Dan-Ling Huang
- grid.263488.30000 0001 0472 9649Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Shuping Zhang
- grid.452223.00000 0004 1757 7615Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zuojun Liu
- grid.263488.30000 0001 0472 9649Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Minxian Qian
- grid.263488.30000 0001 0472 9649Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Ji Li
- grid.452223.00000 0004 1757 7615Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Xian Cheng
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China.
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences; Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China. .,Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University, Shenzhen, China. .,National Engineering Research Center for Biotechnology (Shenzhen); Marshall Laboratory of Biomedical Engineering; International Cancer Center, Shenzhen University, Shenzhen, China. .,Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
18
|
Maiuolo J, Gliozzi M, Carresi C, Musolino V, Oppedisano F, Scarano F, Nucera S, Scicchitano M, Bosco F, Macri R, Ruga S, Cardamone A, Coppoletta A, Mollace A, Cognetti F, Mollace V. Nutraceuticals and Cancer: Potential for Natural Polyphenols. Nutrients 2021; 13:nu13113834. [PMID: 34836091 PMCID: PMC8619660 DOI: 10.3390/nu13113834] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the leading causes of death globally, associated with multifactorial pathophysiological components. In particular, genetic mutations, infection or inflammation, unhealthy eating habits, exposition to radiation, work stress, and/or intake of toxins have been found to contribute to the development and progression of cancer disease states. Early detection of cancer and proper treatment have been found to enhance the chances of survival and healing, but the side effects of anticancer drugs still produce detrimental responses that counteract the benefits of treatment in terms of hospitalization and survival. Recently, several natural bioactive compounds were found to possess anticancer properties, capable of killing transformed or cancerous cells without being toxic to their normal counterparts. This effect occurs when natural products are associated with conventional treatments, thereby suggesting that nutraceutical supplementation may contribute to successful anticancer therapy. This review aims to discuss the current literature on four natural bioactive extracts mostly characterized by a specific polyphenolic profile. In particular, several activities have been reported to contribute to nutraceutical support in anticancer treatment: (1) inhibition of cell proliferation, (2) antioxidant activity, and (3) anti-inflammatory activity. On the other hand, owing to their attenuation of the toxic effect of current anticancer therapies, natural antioxidants may contribute to improving the compliance of patients undergoing anticancer treatment. Thus, nutraceutical supplementation, along with current anticancer drug treatment, may be considered for better responses and compliance in patients with cancer. It should be noted, however, that when data from studies with bioactive plant preparations are discussed, it is appropriate to ensure that experiments have been conducted in accordance with accepted pharmacological research practices so as not to disclose information that is only partially correct.
Collapse
Affiliation(s)
- Jessica Maiuolo
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Oppedisano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Roberta Macri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Annarita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Annachiara Mollace
- Medical Oncology 1, Regina Elena National Cancer Institute, IRCCS, 00144 Rome, Italy; (A.M.); (F.C.)
| | - Francesco Cognetti
- Medical Oncology 1, Regina Elena National Cancer Institute, IRCCS, 00144 Rome, Italy; (A.M.); (F.C.)
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
- Correspondence:
| |
Collapse
|
19
|
Qin T, Zhao J, Liu X, Li L, Zhang X, Shi X, Ke Y, Liu W, Huo J, Dong Y, Shen Y, Li Y, He M, Han S, Li L, Pan C, Wang C. Luteolin combined with low-dose paclitaxel synergistically inhibits epithelial-mesenchymal transition and induces cell apoptosis on esophageal carcinoma in vitro and in vivo. Phytother Res 2021; 35:6228-6240. [PMID: 34494324 DOI: 10.1002/ptr.7267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 08/08/2021] [Accepted: 08/14/2021] [Indexed: 02/05/2023]
Abstract
Although paclitaxel is a promising frontline chemotherapy agent for various malignancies, the clinical applications have been restricted by side effects, drug resistance, and cancer metastasis. The combination of paclitaxel and other agents could be the promising strategies against malignant tumor, which enhances the antitumor effect through synergistic effects, reduces required drug concentrations, and also suppresses tumorigenesis in multiple ways. In this study, we found that luteolin, a natural flavonoid compound, combined with low-dose paclitaxel synergistically regulated the proliferation, migration, epithelial-mesenchymal transition (EMT), and apoptosis of esophageal cancer cells in vitro, as well as synergistically inhibited tumor growth without obvious toxicity in vivo. The molecular mechanism of inhibiting cell migration and EMT processes may be related to the inhibition of SIRT1, and the mechanism of apoptosis induction is associated with the reactive oxygen species (ROS)/c-Jun N-terminal kinase (JNK) pathway-mediated activation of mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Tiantian Qin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Jinzhu Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Xiaojie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Leilei Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Xueyan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Xiaoli Shi
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yu Ke
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Weihua Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Junfeng Huo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Yalong Dong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Yiwei Shen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanyu Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Mingjing He
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shuhua Han
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Linlin Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chengxue Pan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| | - Cong Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and treatment, Zhengzhou, China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, China
| |
Collapse
|
20
|
Qin L, Chen H, Ding X, Guo M, Lang H, Liu J, Li L, Liao J, Liao J. Utilizing network pharmacology to explore potential mechanisms of YiSui NongJian formula in treating myelodysplastic syndrome. Bioengineered 2021; 12:2238-2252. [PMID: 34098848 PMCID: PMC8806438 DOI: 10.1080/21655979.2021.1933867] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The study aims to explore potential mechanisms of YiSui NongJian formula (YSNJF) in treating myelodysplastic syndromes (MDS) by network pharmacology-based strategy. Active compounds and corresponding potential therapeutic targets of YSNJF were harvested by utilizing the database of TCMSP (Traditional Chinese Medicine Systems Pharmacology) and BATMAN-TCM (Bioinformatics Analysis Tool for Molecular mechanism of Traditional Chinese Medicine). MDS targets were adopted from GeneCard, KEGG (Kyoto Encyclopedia of Genes and Genomes), TTD (Therapeutic Target Database), DrugBank, and DisGeNet. Then a network of YSNJF- compounds-target-MDS network was harvested. The protein–protein interaction (PPI) network was then generated by the Sting database and subjected to Cytoscape software to harvest major and core targets network by topological analysis. Genes from the core targets network were further subjected to Gene Ontology (GO) and KEGG enrichment analysis to figure out potential targeting pathways. Finally, a compounds-targets-pathways network was generated by Cytoscape. A total of 210 active compounds and 768 corresponding potential therapeutic targets were harvested from ingredients of YSNJF. MDS was shown to have 772 potential treating targets with 98 intersected targets corresponding to 98 active compounds in YSNJF. Topological analysis revealed that 15 targets formed the core PPI network. Further, GO and KEGG enrichment analysis revealed that those core targets were mainly enriched on cell cycle- and immune-related pathways. The present study revealed that therapeutic effects of YSNJF on MDS might be achieved through regulating cell cycle- and immune-related pathways.
Collapse
Affiliation(s)
- Lerong Qin
- Dongfang Hospital Affiliated, Beijing, China
| | - Haiyan Chen
- Department of Hematology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoqing Ding
- Department of Hematology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ming Guo
- Department of Hematology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Haiyan Lang
- Department of Hematology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junxia Liu
- Department of Hematology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ling Li
- Department of Hematology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Liao
- Department of Hematology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junyao Liao
- Department of Hematology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Nguyen THP, Kumar VB, Ponnusamy VK, Mai TTT, Nhat PT, Brindhadevi K, Pugazhendhi A. Phytochemicals intended for anticancer effects at preclinical levels to clinical practice: Assessment of formulations at nanoscale for non-small cell lung cancer (NSCLC) therapy. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
22
|
Ponte LGS, Pavan ICB, Mancini MCS, da Silva LGS, Morelli AP, Severino MB, Bezerra RMN, Simabuco FM. The Hallmarks of Flavonoids in Cancer. Molecules 2021; 26:2029. [PMID: 33918290 PMCID: PMC8038160 DOI: 10.3390/molecules26072029] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Flavonoids represent an important group of bioactive compounds derived from plant-based foods and beverages with known biological activity in cells. From the modulation of inflammation to the inhibition of cell proliferation, flavonoids have been described as important therapeutic adjuvants against several diseases, including diabetes, arteriosclerosis, neurological disorders, and cancer. Cancer is a complex and multifactor disease that has been studied for years however, its prevention is still one of the best known and efficient factors impacting the epidemiology of the disease. In the molecular and cellular context, some of the mechanisms underlying the oncogenesis and the progression of the disease are understood, known as the hallmarks of cancer. In this text, we review important molecular signaling pathways, including inflammation, immunity, redox metabolism, cell growth, autophagy, apoptosis, and cell cycle, and analyze the known mechanisms of action of flavonoids in cancer. The current literature provides enough evidence supporting that flavonoids may be important adjuvants in cancer therapy, highlighting the importance of healthy and balanced diets to prevent the onset and progression of the disease.
Collapse
Affiliation(s)
- Luis Gustavo Saboia Ponte
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
- Laboratory of Signal Mechanisms (LMS), School of Pharmaceutical Sciences (FCF), University of Campinas (UNICAMP), Campinas, São Paulo 13083-871, Brazil
| | - Mariana Camargo Silva Mancini
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Luiz Guilherme Salvino da Silva
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Ana Paula Morelli
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Matheus Brandemarte Severino
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Rosangela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| |
Collapse
|
23
|
Ganai SA, Sheikh FA, Baba ZA, Mir MA, Mantoo MA, Yatoo MA. Anticancer activity of the plant flavonoid luteolin against preclinical models of various cancers and insights on different signalling mechanisms modulated. Phytother Res 2021; 35:3509-3532. [DOI: 10.1002/ptr.7044] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/04/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Shabir Ahmad Ganai
- Division of Basic Sciences and Humanities FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| | - Farooq Ahmad Sheikh
- Division of Genetics and Plant Breeding FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| | - Zahoor Ahmad Baba
- Division of Basic Sciences and Humanities FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| | - Mudasir Ahmad Mir
- Department of Microbiology Government Medical College Anantnag Jammu & Kashmir India
| | - Mohd Ayoob Mantoo
- Division of Entomology FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| | - Manzoor Ahmad Yatoo
- Division of Basic Sciences and Humanities FoA, SKUAST Kashmir Sopore Jammu & Kashmir India
| |
Collapse
|
24
|
Wang X, Chen B, Xu D, Li Z, Liu H, Huang Z, Huang K, Lin X, Yao H. Molecular mechanism and pharmacokinetics of flavonoids in the treatment of resistant EGF receptor-mutated non-small-cell lung cancer: A narrative review. Br J Pharmacol 2021; 178:1388-1406. [PMID: 33450055 DOI: 10.1111/bph.15360] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/11/2020] [Accepted: 12/09/2020] [Indexed: 01/16/2023] Open
Abstract
Here, we review the molecular mechanism and pharmacokinetics of flavonoids in the treatment of resistant EGF receptor (EGFR)-mutated non-small-cell lung cancer (NSCLC) and particularly the possible mechanism(s) of delicaflavone, a biflavonoid extracted from Selaginella doederleinii Hieron. EGFR TK inhibitors (EGFR-TKI) are ubiquitously used in the treatment of NSCLC bearing EGFR mutations. However, patients treated with EGFR-TKI inevitably and continuously develop resistance. In laboratory studies, flavonoids, as potential adjuvants for cancer chemotherapy, exhibited anti-cancer properties such as inhibition of chemoresistance by interference with ABC transporters-induced drug efflux, curbing of c-MET amplification, or reversal of T790M mutation-mediated resistance. The current review aims at summarizing the association between the anti-cancer potentials of flavonoids and their possible regulatory roles in certain types of mutation that could trigger EGFR-TKI resistance in NSCLC. Potential practical applications of these phytochemicals, as well as the relevant pharmacokinetics, are also discussed.
Collapse
Affiliation(s)
- Xuewen Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Bing Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.,Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou, Fujian, China.,Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou, Fujian, China
| | - Dafen Xu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhijun Li
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Hao Liu
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhengjun Huang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.,Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou, Fujian, China.,Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou, Fujian, China
| | - Kangping Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Xinhua Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.,Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou, Fujian, China.,Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou, Fujian, China
| | - Hong Yao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.,Nano Medical Technology Research Institute, Fujian Medical University, Fuzhou, Fujian, China.,Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
25
|
Li G, Ding K, Qiao Y, Zhang L, Zheng L, Pan T, Zhang L. Flavonoids Regulate Inflammation and Oxidative Stress in Cancer. Molecules 2020; 25:E5628. [PMID: 33265939 PMCID: PMC7729519 DOI: 10.3390/molecules25235628] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer is the second leading cause of death globally. Millions of persons die due to cancer each year. In the last two decades, the anticancer effects of natural flavonoids have become a hot topic in many laboratories. Meanwhile, flavonoids, of which over 8000 molecules are known to date, are potential candidates for the discovery of anticancer drugs. The current review summarizes the major flavonoid classes of anticancer efficacy and discusses the potential anti-cancer mechanisms through inflammation and oxidative stress action, which were based on database and clinical studies within the past years. The results showed that flavonoids could regulate the inflammatory response and oxidative stress of tumor through some anti-inflammatory mechanisms such as NF-κB, so as to realize the anti-tumor effect.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lin Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China; (G.L.); (K.D.); (Y.Q.); (L.Z.); (L.Z.); (T.P.)
| |
Collapse
|
26
|
Autophagy regulation using luteolin: new insight into its anti-tumor activity. Cancer Cell Int 2020; 20:537. [PMID: 33292250 PMCID: PMC7641824 DOI: 10.1186/s12935-020-01634-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
Application of novel methods in cancer therapy is important in terms of management and treatment of the life-threatening disorder. It appears that autophagy is a potential target in cancer therapy, as a variety of drugs targeting autophagy have shown great potential in reducing the viability and proliferation of cancer cells. Autophagy is primarily a catabolic process which provides energy during starvation. Besides, this process contributes to the degradation of aged or potentially toxic components and organelles. On the other hand, the source of a variety of naturally occurring anti-tumor drugs are flavonoids which have high anti-tumor activity. Luteolin is a polyphenolic flavone with the great pharmacological effects such as anti-diabetic, hepatoprotective, antioxidant, anti-inflammation, and anti-tumor. At the present review, we demonstrate how luteolin affects on autophagy process to induce anti-tumor activity.
Collapse
|
27
|
Farooqi AA, Butt G, El-Zahaby SA, Attar R, Sabitaliyevich UY, Jovic JJ, Tang KF, Naureen H, Xu B. Luteolin mediated targeting of protein network and microRNAs in different cancers: Focus on JAK-STAT, NOTCH, mTOR and TRAIL-mediated signaling pathways. Pharmacol Res 2020. [DOI: https://doi.org/10.1016/j.phrs.2020.105188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Farooqi AA, Butt G, El-Zahaby SA, Attar R, Sabitaliyevich UY, Jovic JJ, Tang KF, Naureen H, Xu B. Luteolin mediated targeting of protein network and microRNAs in different cancers: Focus on JAK-STAT, NOTCH, mTOR and TRAIL-mediated signaling pathways. Pharmacol Res 2020; 160:105188. [PMID: 32919041 DOI: 10.1016/j.phrs.2020.105188] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023]
Abstract
There has always been a keen interest of basic and clinical researchers to search for cancer therapeutics having minimum off-target effects and maximum anticancer activities. In accordance with this approach, there has been an explosion in the field of natural products research in the past few decades because of extra-ordinary list of natural extracts and their biologically and pharmacologically active constituents having significant medicinal properties. Apparently, luteolin-mediated anticancer effects have been investigated in different cancers but there is superfluousness of superficial data. Generalized scientific evidence encompassing apoptosis, DNA damage and anti-inflammatory effects has been reported extensively. However, how luteolin modulates deregulated oncogenic pathways in different cancers has not been comprehensively uncovered. In this review we have attempted to focus on cutting-edge research which has unveiled remarkable abilities of luteolin to modulate deregulated oncogenic pathways in different cancers. We have partitioned the review into various sections to separately discuss advancements in therapeutic targeting of oncogenic protein networks. We have provided detailed mechanistic insights related to JAK-STAT signaling and summarized how luteolin inhibited STAT proteins to inhibit STAT-driven gene network. We have also individually analyzed Wnt/β-catenin and NOTCH pathway and how luteolin effectively targeted these pathways. Mapping of the signaling landscape has revealed that NOTCH pathway can be targeted therapeutically. NOTCH pathway was noted to be targeted by luteolin. We have also conceptually analyzed how luteolin restored TRAIL-induced apoptosis in resistant cancers. Luteolin induced an increase in pro-apoptotic proteins and efficiently inhibited anti-apoptotic proteins to induce apoptosis. Luteolin mediated regulation of non-coding RNAs is an exciting and emerging facet. Excitingly, there is sequential and systematic accumulation of clues which have started to shed light on intricate regulation of microRNAs by luteolin in different cancers. Collectively, sophisticated information will enable us to develop a refined understanding of the multi-layered regulation of signaling pathways and non-coding RNAs by luteolin in different cancers.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, 44000, Pakistan.
| | | | - Sally A El-Zahaby
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Rukset Attar
- Department of Obstetrics and Gynecology, Yeditepe University, Turkey
| | - Uteuliyev Yerzhan Sabitaliyevich
- Department of Health Policy and Health Care Development, Kazakh Medical University of Continuing Education, Almaty, 050004, Kazakhstan
| | - Jovana Joksimovic Jovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, SvetozaraMarkovića 69, 34000, Kragujevac, Serbia
| | - Kai-Fu Tang
- Digestive Cancer Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang, China
| | - Humaira Naureen
- Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Baojun Xu
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai, 519087, Guangdong, China.
| |
Collapse
|
29
|
A network pharmacology strategy to investigate the anti-inflammatory mechanism of luteolin combined with in vitro transcriptomics and proteomics. Int Immunopharmacol 2020; 86:106727. [DOI: 10.1016/j.intimp.2020.106727] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/05/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022]
|
30
|
Cao D, Zhu GY, Lu Y, Yang A, Chen D, Huang HJ, Peng SX, Chen LW, Li YW. Luteolin suppresses epithelial-mesenchymal transition and migration of triple-negative breast cancer cells by inhibiting YAP/TAZ activity. Biomed Pharmacother 2020; 129:110462. [PMID: 32768952 DOI: 10.1016/j.biopha.2020.110462] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/21/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly lethal subtype of breast cancer associated with early relapse and metastasis. Epithelial to mesenchymal transition (EMT) plays pivotal roles in the progression of TNBC, including inducing cancer stem cell (CSC) properties, chemoresistance, tumor metastasis, and recurrence. Abnormally activated YAP/TAZ induces EMT in TNBC, making it a promising target for drug development. Our goal is to identify potential YAP/TAZ inhibitors from naturally derivative molecules and further study its effects on inhibiting EMT and metastasis of TNBC. In the current study, we demonstrate that luteolin significantly inhibits YAP/TAZ activity by promoting YAP/TAZ degradation in TNBC cells. Luteolin treatment leads to a decrease of mesenchymal markers and an increase of epithelial markers in both TNBC cells and TAZ-induced mesenchymal cells. Consistently, luteolin treatment inhibits cell migration in TNBC cells. Additionally, luteolin inhibits tumor growth in mice xenografted with TNBC cells. Collectively, our results support luteolin as a novel YAP/TAZ inhibitor for development as a new agent for the treatment of TNBC.
Collapse
Affiliation(s)
- Dai Cao
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guo-Yuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Yan Lu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Aiping Yang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Die Chen
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hui-Jie Huang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shu-Xian Peng
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Li-Wen Chen
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying-Wei Li
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
31
|
Zhuang Z, Chen Q, Huang C, Wen J, Huang H, Liu Z. A Comprehensive Network Pharmacology-Based Strategy to Investigate Multiple Mechanisms of HeChan Tablet on Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:7658342. [PMID: 32595734 PMCID: PMC7277035 DOI: 10.1155/2020/7658342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/03/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND HeChan tablet (HCT) is a traditional Chinese medicine preparation extensively prescribed to treat lung cancer in China. However, the pharmacological mechanisms of HCT on lung cancer remain to be elucidated. METHODS A comprehensive network pharmacology-based strategy was conducted to explore underlying mechanisms of HCT on lung cancer. Putative targets and compounds of HCT were retrieved from TCMSP and BATMAN-TCM databases; related genes of lung cancer were retrieved from OMIM and DisGeNET databases; known therapeutic target genes of lung cancer were retrieved from TTD and DrugBank databases; PPI networks among target genes were constructed to filter hub genes by STRING. Furthermore, the pathway and GO enrichment analysis of hub genes was performed by clusterProfiler, and the clinical significance of hub genes was identified by The Cancer Genome Atlas. RESULT A total of 206 compounds and 2,433 target genes of HCT were obtained. 5,317 related genes of lung cancer and 77 known therapeutic target genes of lung cancer were identified. 507 unique target genes were identified among HCT-related genes of lung cancer and 34 unique target genes were identified among HCT-known therapeutic target genes of lung cancer. By PPI networks, 11 target genes AKT1, TP53, MAPK8, JUN, EGFR, TNF, INS, IL-6, MYC, VEGFA, and MAPK1 were identified as major hub genes. IL-6, JUN, EGFR, and MYC were shown to associate with the survival of lung cancer patients. Five compounds of HCT, quercetin, luteolin, kaempferol, beta-sitosterol, and baicalein were recognized as key compounds of HCT on lung cancer. The gene enrichment analysis implied that HCT probably benefitted patients with lung cancer by modulating the MAPK and PI3K-Akt pathways. CONCLUSION This study predicted pharmacological and molecular mechanisms of HCT against lung cancer and could pave the way for further experimental research and clinical application of HCT.
Collapse
Affiliation(s)
- Zhenjie Zhuang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianying Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cihui Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junmao Wen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haifu Huang
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhanhua Liu
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
32
|
Izzo S, Naponelli V, Bettuzzi S. Flavonoids as Epigenetic Modulators for Prostate Cancer Prevention. Nutrients 2020; 12:E1010. [PMID: 32268584 PMCID: PMC7231128 DOI: 10.3390/nu12041010] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is a multifactorial disease with an unclear etiology. Due to its high prevalence, long latency, and slow progression, PCa is an ideal target for chemoprevention strategies. Many research studies have highlighted the positive effects of natural flavonoids on chronic diseases, including PCa. Different classes of dietary flavonoids exhibit anti-oxidative, anti-inflammatory, anti-mutagenic, anti-aging, cardioprotective, anti-viral/bacterial and anti-carcinogenic properties. We overviewed the most recent evidence of the antitumoral effects exerted by dietary flavonoids, with a special focus on their epigenetic action in PCa. Epigenetic alterations have been identified as key initiating events in several kinds of cancer. Many dietary flavonoids have been found to reverse DNA aberrations that promote neoplastic transformation, particularly for PCa. The epigenetic targets of the actions of flavonoids include oncogenes and tumor suppressor genes, indirectly controlled through the regulation of epigenetic enzymes such as DNA methyltransferase (DNMT), histone acetyltransferase (HAT), and histone deacetylase (HDAC). In addition, flavonoids were found capable of restoring miRNA and lncRNA expression that is altered during diseases. The optimization of the use of flavonoids as natural epigenetic modulators for chemoprevention and as a possible treatment of PCa and other kinds of cancers could represent a promising and valid strategy to inhibit carcinogenesis and fight cancer.
Collapse
Affiliation(s)
- Simona Izzo
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy; (S.I.); (S.B.)
| | - Valeria Naponelli
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy; (S.I.); (S.B.)
- National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d’Oro 305, 00136 Rome, Italy
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, 43124 Parma, Italy
| | - Saverio Bettuzzi
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy; (S.I.); (S.B.)
- National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d’Oro 305, 00136 Rome, Italy
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, 43124 Parma, Italy
| |
Collapse
|
33
|
Costea T, Vlad OC, Miclea LC, Ganea C, Szöllősi J, Mocanu MM. Alleviation of Multidrug Resistance by Flavonoid and Non-Flavonoid Compounds in Breast, Lung, Colorectal and Prostate Cancer. Int J Mol Sci 2020; 21:E401. [PMID: 31936346 PMCID: PMC7013436 DOI: 10.3390/ijms21020401] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the manuscript is to discuss the influence of plant polyphenols in overcoming multidrug resistance in four types of solid cancers (breast, colorectal, lung and prostate cancer). Effective treatment requires the use of multiple toxic chemotherapeutic drugs with different properties and targets. However, a major cause of cancer treatment failure and metastasis is the development of multidrug resistance. Potential mechanisms of multidrug resistance include increase of drug efflux, drug inactivation, detoxification mechanisms, modification of drug target, inhibition of cell death, involvement of cancer stem cells, dysregulation of miRNAs activity, epigenetic variations, imbalance of DNA damage/repair processes, tumor heterogeneity, tumor microenvironment, epithelial to mesenchymal transition and modulation of reactive oxygen species. Taking into consideration that synthetic multidrug resistance agents have failed to demonstrate significant survival benefits in patients with different types of cancer, recent research have focused on beneficial effects of natural compounds. Several phenolic compounds (flavones, phenolcarboxylic acids, ellagitannins, stilbens, lignans, curcumin, etc.) act as chemopreventive agents due to their antioxidant capacity, inhibition of proliferation, survival, angiogenesis, and metastasis, modulation of immune and inflammatory responses or inactivation of pro-carcinogens. Moreover, preclinical and clinical studies revealed that these compounds prevent multidrug resistance in cancer by modulating different pathways. Additional research is needed regarding the role of phenolic compounds in the prevention of multidrug resistance in different types of cancer.
Collapse
Affiliation(s)
- Teodora Costea
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Oana Cezara Vlad
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| | - Luminita-Claudia Miclea
- Department of Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Research Excellence Center in Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Constanta Ganea
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Maria-Magdalena Mocanu
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| |
Collapse
|
34
|
Hazafa A, Rehman KU, Jahan N, Jabeen Z. The Role of Polyphenol (Flavonoids) Compounds in the Treatment of Cancer Cells. Nutr Cancer 2020; 72:386-397. [PMID: 31287738 DOI: 10.1080/01635581.2019.1637006] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer remains a second leading cause of deaths and major public health problem. It occurs due to extensive DNA damage caused by ultraviolet radiations, ionizing radiations, environmental agents, therapeutic agents, etc. Among all cancers, the most frequently diagnosed cancers are lung (12.7%), breast (10.9%), colorectal (9.7%), and gastric cancer (7.81%). Natural compounds are most favorable against cancer on the count of their anti-cancerous ability, easy to avail and efficient. Among natural compounds, polyphenols (flavonoids, catechin, hesperetin, flavones, quercetin, phenolic acids, ellagic acid, lignans, stilbenes, etc.) represent a large and diverse group used in the prevention and treatment of cancer. Natural flavonoids are derived from different plant sources and from various medicinal plants including Petroselinum crispum, Apium graveolens, Flemingia vestita, Phyllanthus emblica, etc. Natural flavonoids possess antioxidant, anti-inflammation, as well as anti-cancerous activities through multiple pathways, they induce apoptosis in breast, colorectal, and prostate cancers, lower the nucleoside diphosphate kinase-B activity in lung, bladder and colon cancers, inhibit cell-proliferation and cell cycle arrest by suppressing the NF-kB pathway in various cancers, etc. The current review summarized the anticancer activities of natural polyphenols and their mechanisms of action.
Collapse
Affiliation(s)
- Abu Hazafa
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Khalil-Ur- Rehman
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Nazish Jahan
- Department of Chemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Zara Jabeen
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
35
|
Ma Y, Ma M, Ma L, Zhang F, Liu Y, Ma X. Downregulation of miR-552 in hepatocellular carcinoma inhibits cell migration and invasion, and promotes cell apoptosis via RUNX3. Exp Ther Med 2019; 18:3829-3836. [PMID: 31656538 PMCID: PMC6812473 DOI: 10.3892/etm.2019.8061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 08/07/2019] [Indexed: 12/27/2022] Open
Abstract
Research conducted previously has indicated that microRNAs (miRs) have potential effects on the pathogenesis of hepatocellular carcinoma (HCC). The biological functions of miR-552 have been well documented in colon cancer; however, the role of miR-552 in HCC remains unclear. The present study evaluated the effects of miR-552 in HCC physiology, using HCC cell lines as model. An miR-552 inhibitor was transfected into HCC cell lines to knock down the expression of miR-552. Reverse transcription-quantitative PCR and western blot analysis were used to detect the expression of miR-552 and Runt-related transcription factor 3 (RUNX3), respectively. MTT assay was used to analyze cell viability, whilst Transwell and wound-healing assay were used to investigate cell migration. Flow cytometry was performed to measure cell apoptosis. The direct association between RUNX3 and miR-552 was evaluated using dual luciferase reporter assay. The expression of miR-552 was significantly elevated in HCC tumor tissues compared with the adjacent healthy samples. Additionally, transfection with the miR-552 inhibitor decreased cell viability and migration. miR-552 knockdown also increased HCC cell apoptosis in vitro. In conclusion, these results suggest that miR-552 has an oncogenic function in HCC and is a potential biomarker for detecting HCC.
Collapse
Affiliation(s)
- Ying Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang 830054, P.R. China
| | - Ming Ma
- Department of Hepatobiliary Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumchi, Xinjiang 830001, P.R. China
| | - Ling Ma
- Department of Pharmacy, Thoracic Hospital of Xinjiang Uygur Autonomous Region, Urumchi, Xinjiang 830049, P.R. China
| | - Fengbo Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang 830054, P.R. China
| | - Yumei Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang 830054, P.R. China
| | - Xiumin Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang 830054, P.R. China
| |
Collapse
|
36
|
Ma B, Zhang J, Zhu Z, Zhao A, Zhou Y, Ying H, Zhang Q. Luteolin Ameliorates Testis Injury and Blood–Testis Barrier Disruption through the Nrf2 Signaling Pathway and by Upregulating Cx43. Mol Nutr Food Res 2019; 63:e1800843. [DOI: 10.1002/mnfr.201800843] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 02/21/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Bo Ma
- School of Pharmaceutical SciencesNanjing Tech University Nanjing 210009 P. R. China
| | - Jie Zhang
- School of Pharmaceutical SciencesNanjing Tech University Nanjing 210009 P. R. China
| | - Zhiming Zhu
- School of Pharmaceutical SciencesNanjing Tech University Nanjing 210009 P. R. China
| | - Ang Zhao
- School of Pharmaceutical SciencesNanjing Tech University Nanjing 210009 P. R. China
| | - Yanfen Zhou
- School of Pharmaceutical SciencesNanjing Tech University Nanjing 210009 P. R. China
| | - Hanjie Ying
- School of Life Science & Pharmaceutical EngineeringNanjing University of Technology Nanjing 210009 P. R. China
| | - Qi Zhang
- School of Pharmaceutical SciencesNanjing Tech University Nanjing 210009 P. R. China
| |
Collapse
|
37
|
Imran M, Rauf A, Abu-Izneid T, Nadeem M, Shariati MA, Khan IA, Imran A, Orhan IE, Rizwan M, Atif M, Gondal TA, Mubarak MS. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed Pharmacother 2019; 112:108612. [PMID: 30798142 DOI: 10.1016/j.biopha.2019.108612] [Citation(s) in RCA: 487] [Impact Index Per Article: 81.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/06/2019] [Accepted: 01/23/2019] [Indexed: 10/27/2022] Open
Abstract
Many food-derived phytochemicals and their derivatives represent a cornucopia of new anti-cancer compounds. Luteolin (3,4,5,7-tetrahydroxy flavone) is a flavonoid found in different plants such as vegetables, medicinal herbs, and fruits. It acts as an anticancer agent against various types of human malignancies such as lung, breast, glioblastoma, prostate, colon, and pancreatic cancers. It also blocks cancer development in vitro and in vivo by inhibition of proliferation of tumor cells, protection from carcinogenic stimuli, and activation of cell cycle arrest, and by inducing apoptosis through different signaling pathways. Luteolin can additionally reverse epithelial-mesenchymal transition (EMT) through a mechanism that involves cytoskeleton shrinkage, induction of the epithelial biomarker E-cadherin expression, and by down-regulation of the mesenchymal biomarkers N-cadherin, snail, and vimentin. Furthermore, luteolin increases levels of intracellular reactive oxygen species (ROS) by activation of lethal endoplasmic reticulum stress response and mitochondrial dysfunction in glioblastoma cells, and by activation of ER stress-associated proteins expressions, including phosphorylation of eIF2α, PERK, CHOP, ATF4, and cleaved-caspase 12. Accordingly, the present review article summarizes the progress of recent research on luteolin against several human cancers.
Collapse
Affiliation(s)
- Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahor, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, 23561, Khyber Pakhtunkhwa, Pakistan.
| | - Tareq Abu-Izneid
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University of Science and Technology, Al Ain Campus, UAE
| | - Muhammad Nadeem
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Vehari, Pakistan
| | - Mohammad Ali Shariati
- Laboratory of Biocontrol and Antimicrobial Resistance, Orel State, University Named After I.S. Turgenev, 302026, Orel, Russia
| | - Imtiaz Ali Khan
- Department of Agriculture, University of Swabi, Anbar, 23561, Khyber Pakhtunkhwa, Pakistan
| | - Ali Imran
- Department of Food Science, Nutrition & Home Economics, Institute of Home and Food Sciences, Government College University, Faisalabad, Pakistan
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey.
| | - Muhammad Rizwan
- Department of Microbiology and Biotechnology, Abasyn University Peshawar, KPK, Pakistan
| | - Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Faculty of Health, Deakin University, Australia
| | - Mohammad S Mubarak
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
38
|
Ryu S, Park S, Lim W, Song G. Effects of luteolin on canine osteosarcoma: Suppression of cell proliferation and synergy with cisplatin. J Cell Physiol 2018; 234:9504-9514. [DOI: 10.1002/jcp.27638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/02/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Soomin Ryu
- Department of Biotechnology Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University Seoul Republic of Korea
| | - Sunwoo Park
- Department of Biotechnology Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University Seoul Republic of Korea
| | - Whasun Lim
- Department of Biomedical Sciences Catholic Kwandong University Gangneung Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University Seoul Republic of Korea
| |
Collapse
|
39
|
Anson DM, Wilcox RM, Huseman ED, Stump TA, Paris RL, Darkwah BO, Lin S, Adegoke AO, Gryka RJ, Jean-Louis DS, Amos S. Luteolin Decreases Epidermal Growth Factor Receptor-Mediated Cell Proliferation and Induces Apoptosis in Glioblastoma Cell Lines. Basic Clin Pharmacol Toxicol 2018; 123:678-686. [PMID: 29935053 DOI: 10.1111/bcpt.13077] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022]
Abstract
Glioblastomas are a subtype of gliomas, which are the most aggressive and deadly form of brain tumours. The epidermal growth factor receptor (EGFR) is over-expressed and amplified in glioblastomas. Luteolin is a common bioflavonoid found in a variety of fruits and vegetables. The aim of this study was to explore the molecular and biological effects of luteolin on EGF-induced cell proliferation and the potential of luteolin to induce apoptosis in glioblastoma cells. In vitro cell viability assays demonstrated that luteolin decreased cell proliferation in the presence or absence of EGF. Immunoblots revealed that luteolin decreased the protein expression levels of phosphorylated Akt, mTOR, p70S6K and MAPK in the presence of EGF. Furthermore, our results revealed the ability of luteolin to induce caspase and PARP cleavages in glioblastoma cells in addition to promoting cell cycle arrest. Our results demonstrated that luteolin has an inhibitory effect on downstream signalling molecules activated by EGFR, particularly the Akt and MAPK signalling pathways, and provided a rationale for further clinical investigation into the use of luteolin as a therapeutic molecule in the management of glioblastoma.
Collapse
Affiliation(s)
- David M Anson
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Rachel M Wilcox
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Eric D Huseman
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Trevor A Stump
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Robert L Paris
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Belinda O Darkwah
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Stacy Lin
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Andrea O Adegoke
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Rebecca J Gryka
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Denise S Jean-Louis
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| | - Samson Amos
- Department of Pharmaceutical Sciences, School of Pharmacy, Cedarville University, Cedarville, OH, USA
| |
Collapse
|
40
|
Cao X, Wang M, Sun L, Ren X, Pei G. Preferential adsorption of flavonoids from peanut shell by amino-modified Fe 3 O 4 nanoparticles (MNP-NH 2 ). JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:3588-3594. [PMID: 29315633 DOI: 10.1002/jsfa.8875] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/26/2017] [Accepted: 12/29/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND The highly selective capture of individual components of complex systems such as Chinese medicine extracts is a great challenge. With the rapid development of nanoscience, magnetic materials have attracted increased attention as suitable candidates for use in drug transport vehicles. RESULTS In this work, amino-modified Fe3 O4 nanoparticles (MNP-NH2 ) have been synthesized and successfully used to selectively isolate luteolin, a type of flavonoid, from peanut shell, coupled with high-performance liquid chromatography. MNP-NH2 were characterized by transmission electron microscopy, scanning electron microscopy, Fourier transform infrared spectroscopy and vibrating sample magnetometer analysis, which showed that the amino functional groups were successfully bound to the surface of the magnetic particles. Adsorption of six different flavonoids by MNP-NH2 showed that the mechanism of adsorption was related to the number of ortho-phenolic hydroxyl groups. The optimized adsorption conditions were extraction for 40 min at 30 °C by single-factor experiments and the final elution conditions were determined to be 5 mL of 20% glacial acetic acid (methanol:water = 60:40) and ultrasonication for 40 min by L9 (34 ) orthogonal test. CONCLUSION Based on these findings, we first created a method for separating luteolin from peanut shell extract by MNP-NH2 . This newly developed magnetic method of extracting chemical compounds from herbal foodstuffs and medicines provides new ideas for the utilization of a cash crop. © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xuexiao Cao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Meng Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lili Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Gaosheng Pei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
41
|
Huang X, Bhugul PA, Fan G, Ye T, Huang S, Dai S, Chen B, Zhou M. Luteolin inhibits pancreatitis‑induced acinar‑ductal metaplasia, proliferation and epithelial‑mesenchymal transition of acinar cells. Mol Med Rep 2017; 17:3681-3689. [PMID: 29286098 PMCID: PMC5802174 DOI: 10.3892/mmr.2017.8327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 07/14/2017] [Indexed: 01/05/2023] Open
Abstract
Luteolin, a flavone, has been demonstrated to have anti‑cancer properties. In the current study, the effects of luteolin on certain carcinogenesis‑associated changes induced by pancreatitis, which are significant risk factors for pancreatic cancer, were investigated. Male six‑week‑old C57BL6 mice used in the current study were divided into three groups; the control group, acute pancreatitis group and luteolin group. Intra‑peritoneal injection of cearulein was performed in the acute pancreatitis group and luteolin group to induce acute pancreatitis whereas the luteolin group received intra‑peritoneal injection of luteolin. The control group received intra‑peritoneal injection of normal saline. Then, the expression of SOX9, phosphorylated (p‑) STAT3, p‑EGFR, cytokeratin‑19, Ki67 and N‑cadherin were determined by immunohistochemistry. Morphological changes of acinar cells were determined by hematoxylin and eosin staining. The mRNA expression of the epithelial‑mesenchymal transition markers CDH1, CDH2, Slug, Zeb1, EpCAM, ZO1, Vimentin, Snail and Twist was determined by reverse transcription‑quantitative polymerase chain reaction. It was identified that luteolin inhibits the formation of tubular complexes and ectopic expression of cytokeratin‑19 and luteolin also decreased proteins of SOX9, p‑STAT3 and p‑EGFR. In addition, luteolin inhibits proliferation and epithelial‑mesenchymal transition of acinar cells induced by acute pancreatitis. As tubular complex formation and ectopic expression of cytokeratin‑19 were two prominent characters of acinar‑ductal metaplasia, it was concluded that luteolin inhibits acinar‑ductal metaplasia induced by pancreatitis and also inhibits pancreatitis‑induced proliferation and epithelial‑mesenchymal transition of acinar cells. Acinar‑ductal metaplasia and proliferation have close associations with pancreatic carcinogenesis. It is suggested that luteolin has potential anti‑pancreatic carcinogenesis effects and merits further investigation.
Collapse
Affiliation(s)
- Xince Huang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Pravin Avinash Bhugul
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Gang Fan
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Tingting Ye
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Shihao Huang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Shengjie Dai
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Bicheng Chen
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Mengtao Zhou
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
42
|
Suvarna V, Murahari M, Khan T, Chaubey P, Sangave P. Phytochemicals and PI3K Inhibitors in Cancer-An Insight. Front Pharmacol 2017; 8:916. [PMID: 29311925 PMCID: PMC5736021 DOI: 10.3389/fphar.2017.00916] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/30/2017] [Indexed: 12/11/2022] Open
Abstract
In today's world of modern medicine and novel therapies, cancer still remains to be one of the prime contributor to the death of people worldwide. The modern therapies improve condition of cancer patients and are effective in early stages of cancer but the advanced metastasized stage of cancer remains untreatable. Also most of the cancer therapies are expensive and are associated with adverse side effects. Thus, considering the current status of cancer treatment there is scope to search for efficient therapies which are cost-effective and are associated with lesser and milder side effects. Phytochemicals have been utilized for many decades to prevent and cure various ailments and current evidences indicate use of phytochemicals as an effective treatment for cancer. Hyperactivation of phosphoinositide 3-kinase (PI3K) signaling cascades is a common phenomenon in most types of cancers. Thus, natural substances targeting PI3K pathway can be of great therapeutic potential in the treatment of cancer patients. This chapter summarizes the updated research on plant-derived substances targeting PI3K pathway and the current status of their preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Vasanti Suvarna
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Manikanta Murahari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S Ramaiah University of Applied Sciences, Bangalore, India
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Pramila Chaubey
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Preeti Sangave
- Department of Pharmaceutical Sciences, School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, India
| |
Collapse
|
43
|
Li J, Cheng X, Chen Y, He W, Ni L, Xiong P, Wei M. Vitamin E TPGS modified liposomes enhance cellular uptake and targeted delivery of luteolin: An in vivo/in vitro evaluation. Int J Pharm 2016; 512:262-272. [DOI: 10.1016/j.ijpharm.2016.08.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 08/01/2016] [Accepted: 08/17/2016] [Indexed: 01/12/2023]
|
44
|
Natural Polyphenols for Prevention and Treatment of Cancer. Nutrients 2016; 8:nu8080515. [PMID: 27556486 PMCID: PMC4997428 DOI: 10.3390/nu8080515] [Citation(s) in RCA: 428] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/12/2016] [Accepted: 08/12/2016] [Indexed: 02/06/2023] Open
Abstract
There is much epidemiological evidence that a diet rich in fruits and vegetables could lower the risk of certain cancers. The effect has been attributed, in part, to natural polyphenols. Besides, numerous studies have demonstrated that natural polyphenols could be used for the prevention and treatment of cancer. Potential mechanisms included antioxidant, anti-inflammation as well as the modulation of multiple molecular events involved in carcinogenesis. The current review summarized the anticancer efficacy of major polyphenol classes (flavonoids, phenolic acids, lignans and stilbenes) and discussed the potential mechanisms of action, which were based on epidemiological, in vitro, in vivo and clinical studies within the past five years.
Collapse
|
45
|
Pratheeshkumar P, Son YO, Divya SP, Wang L, Turcios L, Roy RV, Hitron JA, Kim D, Dai J, Asha P, Zhang Z, Shi X. Quercetin inhibits Cr(VI)-induced malignant cell transformation by targeting miR-21-PDCD4 signaling pathway. Oncotarget 2016; 8:52118-52131. [PMID: 28881718 PMCID: PMC5581017 DOI: 10.18632/oncotarget.10130] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 06/03/2016] [Indexed: 12/16/2022] Open
Abstract
Hexavalent chromium [Cr(VI)] is an important human carcinogen associated with pulmonary diseases and lung cancer. Inhibition of Cr(VI)-induced carcinogenesis by a dietary antioxidant is a novel approach. Quercetin is one of the most abundant dietary flavonoids widely present in many fruits and vegetables, possesses potent antioxidant and anticancer properties. MicroRNA-21 (miR-21) is a key oncomiR significantly elevated in the majority of human cancers that exerts its oncogenic activity by targeting the tumor suppressor gene programmed cell death 4 (PDCD4). The present study examined the effect of quercetin on the inhibition of Cr(VI)-induced malignant cell transformation and the role of miR-21-PDCD4 signaling involved. Our results showed that quercetin decreased ROS generation induced by Cr(VI) exposure in BEAS-2B cells. Chronic Cr(VI) exposure induced malignant cell transformation, increased miR-21 expression and caused inhibition of PDCD4, which were significantly inhibited by the treatment of quercetin in a dose dependent manner. Nude mice injected with BEAS-2B cells chronically exposed to Cr(VI) in the presence of quercetin showed reduced tumor incidence compared to Cr(VI) alone treated group. Stable knockdown of miR-21 and overexpression of PDCD4 or catalase in BEAS-2B cells suppressed Cr(VI)-induced malignant transformation and tumorigenesis. Taken together, these results demonstrate that quercetin is able to protect BEAS-2B cells from Cr(VI)-induced carcinogenesis by targeting miR-21-PDCD4 signaling.
Collapse
Affiliation(s)
- Poyil Pratheeshkumar
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Young-Ok Son
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Sasidharan Padmaja Divya
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Lei Wang
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Lilia Turcios
- Department of Surgery, University of Kentucky, College of Medicine, Lexington, KY, USA
| | - Ram Vinod Roy
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - John Andrew Hitron
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Donghern Kim
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Jin Dai
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Padmaja Asha
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Cochin, India
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Xianglin Shi
- Center for Research on Environmental Disease, University of Kentucky, Lexington, KY, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
46
|
Tjioe KC, Tostes Oliveira D, Gavard J. Luteolin Impacts on the DNA Damage Pathway in Oral Squamous Cell Carcinoma. Nutr Cancer 2016; 68:838-47. [DOI: 10.1080/01635581.2016.1180411] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Han K, Meng W, Zhang JJ, Zhou Y, Wang YL, Su Y, Lin SC, Gan ZH, Sun YN, Min DL. Luteolin inhibited proliferation and induced apoptosis of prostate cancer cells through miR-301. Onco Targets Ther 2016; 9:3085-94. [PMID: 27307749 PMCID: PMC4888721 DOI: 10.2147/ott.s102862] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Luteolin is a falvonoid compound derived from Lonicera japonica Thunb. Numerous reports have demonstrated that luteolin has anticancer effects on many kinds of tumors. This study investigated the effects of luteolin on prostate cancer (PCa), assessing the PC3 and LNCaP cells. The cell viability and apoptosis were assessed by performing Cell Counting Kit-8 assay and Annexin V–fluorescein isothiocyanate/propidium iodide double staining. Luteolin was found to inhibit androgen-sensitive and androgen-independent PCa cell lines’ growth and induced apoptosis. To uncover the exact mechanisms and molecular targets, microRNA (miR) array analysis was performed. miR-301 was found to be markedly downregulated. Then, the expression of miR-301 was retrospectively analyzed in the primary PCa tissues by quantitative reverse transcription polymerase chain reaction and in situ hybridization methods. According to the quantitative reverse transcription polymerase chain reaction results of miR-301, the 54 PCa patients were divided into two groups: high and low miR-301 groups. The division indicator is a relative expression ≥5. Compared to the low-expression group, high miR-301 expression was associated with a significantly shorter overall survival (P=0.029). The proapoptotic gene, DEDD2, was predicted to be the direct target of miR-301. It was clarified in accordance with bioinformatics and luciferase activity analyses. The overexpression of miR-301 by plasmid decreased the luteolin effect. Taken together, these results suggest that luteolin inhibits PCa cell proliferation through miR-301, the poor predictive factor of PCa.
Collapse
Affiliation(s)
- Kun Han
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Wei Meng
- Institute of Genetic Engineering of Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jian-Jun Zhang
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Yan Zhou
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Ya-Ling Wang
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Yang Su
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Shu-Chen Lin
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Zhi-Hua Gan
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Yong-Ning Sun
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Da-Liu Min
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| |
Collapse
|
48
|
Huang X, Dai S, Dai J, Xiao Y, Bai Y, Chen B, Zhou M. Luteolin decreases invasiveness, deactivates STAT3 signaling, and reverses interleukin-6 induced epithelial-mesenchymal transition and matrix metalloproteinase secretion of pancreatic cancer cells. Onco Targets Ther 2015; 8:2989-3001. [PMID: 26527884 PMCID: PMC4621199 DOI: 10.2147/ott.s91511] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Luteolin, a flavone, has been shown to exhibit anticancer properties. Here, we investigated whether luteolin affects epithelial–mesenchymal transition (EMT) and invasiveness of pancreatic cancer cell lines and their underlying mechanism. Pancreatic cancer cell lines PANC-1 and SW1990 were used in our study, and their EMT characters, matrix metalloproteinase (MMP) expression level, invasiveness, and signal transducer and activator of transcription 3 (STAT3) activity were determined after luteolin treatment. We also treated pancreatic cancer cells with interleukin-6 (IL-6) to see whether IL-6-induced activation of STAT3, EMT, and MMP secretion was affected by luteolin. We found that luteolin inhibits EMT and MMP2, MMP7, and MMP9 expression in a dose-dependent manner, similar to STAT3 signaling. Through Transwell assay, we found that invasiveness of pancreatic cancer cells was inhibited by luteolin. EMT characters and MMP secretion increase with STAT3 activity after IL-6 treatment and these effects, caused by IL-6, were inhibited by luteolin. We concluded that luteolin inhibits invasiveness of pancreatic cancer cells, and we speculated that luteolin inhibits EMT and MMP secretion likely through deactivation of STAT3 signaling. Luteolin has potential antitumor effects and merits further investigation.
Collapse
Affiliation(s)
- Xince Huang
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Shengjie Dai
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Juji Dai
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Yuwu Xiao
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Yongyu Bai
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| | - Bicheng Chen
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China ; Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Wenzhou, Zhejiang Province, People's Republic of China
| | - Mengtao Zhou
- Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
49
|
Huang L, Fu L. Mechanisms of resistance to EGFR tyrosine kinase inhibitors. Acta Pharm Sin B 2015; 5:390-401. [PMID: 26579470 PMCID: PMC4629442 DOI: 10.1016/j.apsb.2015.07.001] [Citation(s) in RCA: 363] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/12/2015] [Accepted: 06/13/2015] [Indexed: 12/30/2022] Open
Abstract
Since the discovery that non-small cell lung cancer (NSCLC) is driven by epidermal growth factor receptor (EGFR) mutations, the EGFR tyrosine kinase inhibitors (EGFR-TKIs, e.g., gefitinib and elrotinib) have been effectively used for clinical treatment. However, patients eventually develop drug resistance. Resistance to EGFR-TKIs is inevitable due to various mechanisms, such as the secondary mutation (T790M), activation of alternative pathways (c-Met, HGF, AXL), aberrance of the downstream pathways (K-RAS mutations, loss of PTEN), impairment of the EGFR-TKIs-mediated apoptosis pathway (BCL2-like 11/BIM deletion polymorphism), histologic transformation, ATP binding cassette (ABC) transporter effusion, etc. Here we review and summarize the known resistant mechanisms to EGFR-TKIs and provide potential targets for development of new therapeutic strategies.
Collapse
Key Words
- ABC, ATP binding cassette
- ABCB1, ATP binding cassette, sub-family B, member 1
- ABCC1, ATP binding cassette, sub-family C, member 1
- ABCC10, ATP binding cassette, sub-family C, member 10
- ABCG2, ATP binding cassette, sub-family G, member 2
- AKT, protein kinase B
- ALK, anaplastic lymphoma kinase
- AXL, Anexelekto
- BCL-2, B-cell CLL/lymphoma-2
- BCL2L11/BIM, BCL2-like 11
- BH3, BCL2-homology domain 3
- BRAF, v-RAF murine sarcoma viral oncogene homolog B1
- CML, chronic myelogenous leukemia
- CRKL, Crk-like protein
- EGFR
- EGFR, epidermal growth factor receptor
- EGFR-TKIs, epidermal growth factor receptor tyrosine kinase inhibitors
- EGFRvIII, EGFR variant III
- EML4, echinoderm microtubule-associated protein-like 4
- EMT, epithelial mesenchymal transition
- ERK1/2, extracellular signal-regulated kinases
- FGFRs, fibroblast growth factor receptors
- FGFs, fibroblast growth factors
- GAS6, growth-arrest-specific protein 6
- HER, human epidermal receptor
- HGF, hepatocyte growth factor
- IGF, insulin growth factor
- IGF-1R, IGF-1 receptor
- IGFBPs, IGF-binding proteins
- IL, interleukin
- IL-6R, IL-6 receptor
- JAK, janus kinase
- MAPK, mitogen-activated protein kinase
- MEK, mitogen-activated protein kinase
- Mechanisms
- NSCLC, non-small cell lung cancer
- PDGFRs, platelet-derived growth factor receptors
- PDGFs, platelet-derived growth factors
- PI3K, phosphatidylinositol-3-kinase
- PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase,catalytic subunit alpha
- PTEN, phosphatase and tensin homolog
- RAF, rapidly accelerated fibrosarcoma
- RAS, rat sarcoma
- RTK, tyrosine kinase receptor
- Resistance
- SF, scatter factor
- SOCS3, suppressor of cytokine signaling 3
- STAT, signal transducers and activators of transcription
- TKIs
- TKIs, tyrosine kinase inhibitors
- TKs, tyrosine kinases
- VEGF, vascular endothelial growth factor
- VEGFR, vascular endothelial growth factor receptor
Collapse
Affiliation(s)
| | - Liwu Fu
- Corresponding author. Tel.: +86 20 87343163; fax: +86 20 87343170.
| |
Collapse
|
50
|
Wang Q, Zhang J, Liu M, Yang J, Zhang XM, Zhou L, Cao L, Liao XL. Modified Syntheses of the Dietary Flavonoid Luteolin. JOURNAL OF CHEMICAL RESEARCH 2015. [DOI: 10.3184/174751915x14404221529907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Two novel syntheses of the flavone luteolin are described. In the first, 3,5-dimethoxyphenol was converted to 2-hydroxy-4,6-dimethoxyacetophenone and then by condensation with 3,4-dimethoxybenzaldehyde to 2′-hydroxy-3,4,4′,6′-tetramethoxychalcone. In the second, the chalcone step was prepared in which 3,5-dimethoxyphenol was acylated with 3,4-dimethoxycinnamoyl chloride. The chalcone was then cyclised with iodine and demethylated with pyridine hydrochloride to form luteolin in 47% and 40% overall yield, respectively. Several disadvantages of previous syntheses like long reaction time, harsh reaction conditions and low overall yield have been overcome.
Collapse
Affiliation(s)
- Qian Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 South Jingming Road, Chenggong District, Kunming, Yunnan Province 650500, P.R. China
| | - Ji Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 South Jingming Road, Chenggong District, Kunming, Yunnan Province 650500, P.R. China
| | - Man Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 South Jingming Road, Chenggong District, Kunming, Yunnan Province 650500, P.R. China
| | - Jian Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 South Jingming Road, Chenggong District, Kunming, Yunnan Province 650500, P.R. China
| | - Xiang-ming Zhang
- College of Pharmacy, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, P.R. China
- Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, Tianjin Economic and Technological Development Zone, Tianjin 300457, P.R. China
| | - Lei Zhou
- College of Pharmacy, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, P.R. China
- Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, Tianjin Economic and Technological Development Zone, Tianjin 300457, P.R. China
| | - Lang Cao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 South Jingming Road, Chenggong District, Kunming, Yunnan Province 650500, P.R. China
| | - Xia-li Liao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, 727 South Jingming Road, Chenggong District, Kunming, Yunnan Province 650500, P.R. China
| |
Collapse
|