1
|
Helen H, Gunawan MC, Halim P, Dinata MR, Ahmed A, Dalimunthe A, Marianne M, Ribeiro RIMDA, Hasibuan PAZ, Nurkolis F, Hey-Hawkins E, Park MN, Harahap U, Kim SH, Kim B, Syahputra RA. Flavonoids as modulators of miRNA expression in pancreatic cancer: Pathways, Mechanisms, And Therapeutic Potential. Biomed Pharmacother 2024; 179:117347. [PMID: 39241569 DOI: 10.1016/j.biopha.2024.117347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024] Open
Abstract
Pancreatic cancer (PC) is a complex malignancy, distinguished by its aggressive characteristics and unfavorable prognosis. Recent developments in understanding the molecular foundations of this disease have brought attention to the noteworthy involvement of microRNAs (miRNAs) in disease development, advancement, and treatment resistance. The anticancer capabilities of flavonoids, which are a wide range of phytochemicals present in fruits and vegetables, have attracted considerable interest because of their ability to regulate miRNA expression. This review provides the effects of flavonoids on miRNA expression in PC, explains the underlying processes, and explores the possible therapeutic benefits of flavonoid-based therapies. Flavonoids inhibit PC cell proliferation, induce apoptosis, and enhance chemosensitivity via the modulation of miRNAs involved in carcinogenesis. Additionally, this review emphasizes the significance of certain miRNAs as targets of flavonoid action. These miRNAs have a role in regulating important signaling pathways such as the phosphoinositide-3-kinase-protein kinase B/Protein kinase B (Akt), mitogen activated protein kinase (MAPK), Janus kinase/signal transducers and activators of transcription (JAK/STAT), and Wnt/β-catenin pathways. This review aims to consolidate current knowledge on the interaction between flavonoids and miRNAs in PC, providing a comprehensive analysis of how flavonoid-mediated modulation of miRNA expression could influence cancer progression and therapy. It highlights the use of flavonoid nanoformulations to enhance stability, increase absorption, and maximize anti-PC activity, improving patient outcomes. The review calls for further research to optimize the use of flavonoid nanoformulations in clinical trials, leading to innovative treatment strategies and more effective approaches for PC.
Collapse
Affiliation(s)
- Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Mega Carensia Gunawan
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Muhammad Riza Dinata
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Amer Ahmed
- Department of Bioscience, Biotechnology and Environment, University of Bari, Bari, Italy
| | - Aminah Dalimunthe
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Marianne Marianne
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Rosy Iara Maciel De Azambuja Ribeiro
- Experimental Pathology Laboratory, Federal University of São João del Rei (UFSJ), 400, Sebastião Gonçalves Coelho, Chanadour, Divinópolis 35501-296, MG, Brazil
| | | | - Fahrul Nurkolis
- Biological Sciences, Faculty of Sciences and Technology, UIN Sunan Kalijaga, Yogyakarta, Indonesia
| | - Evamarie Hey-Hawkins
- Leipzig University, Faculty of Chemistry and Mineralogy, Centre for Biotechnology and Biomedicine (BBZ), Institute of Bioanalytical Chemistry, Deutscher Platz 5, 04103 Leipzig, Germany
| | - Moon Nyeo Park
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; College of Korean Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Urip Harahap
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia
| | - Sung-Hoon Kim
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Bonglee Kim
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; College of Korean Medicine, Kyung Hee University, Hoegidong, Dongdaemungu, Seoul, 05253, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Sumatera Utara, Indonesia.
| |
Collapse
|
2
|
Martynyuk VA, Efimova SS, Malykhina AI, Ostroumova OS. The effects of plant flavones on the membrane boundary potential and lipid packing stress. Colloids Surf B Biointerfaces 2024; 245:114269. [PMID: 39341052 DOI: 10.1016/j.colsurfb.2024.114269] [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: 03/23/2024] [Revised: 08/12/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024]
Abstract
Here we have revealed the effects of different plant flavones on the physicochemical properties of model lipid membranes. We have demonstrated that baicalein increases the boundary potential of membranes composed of phosphatidylcholine, while wogonin does not affect it. Other flavones tested reduce membrane boundary potential, with this ability increasing among scutellarein, chrysin, apigenin, morin, fisetin, and luteolin. Molecular dynamics simulations demonstrate connection of alteration in boundary potential with the preferential orientation of intrinsic flavone dipole moments in membranes. We have also shown that flavones reduce the melting point of phosphatidylcholine, and this ability increases in the series of luteolin, morin, wogonin, scutellarein, apigenin, baicalein, chrysin, and fisetin. The introduction of baicalein, chrysin and fisetin also leads to a significant decrease in the sharpness of the lipid phase transition. We have hypothesized that the localization of flavones in the glycerol backbone or in the C1-C8 methylene region of lipid hydrocarbon chains leads to an increase in the area per lipid and, as a consequence, to an expansion of the lipid melting peak. Replacement of neutral phosphatidylcholine with negatively charged phosphatidylserine affects the membrane-modifying activity of flavones which given the externalization of phosphatidylserine on the surface of cancer cells may be crucial in the flavone anticancer effects.
Collapse
Affiliation(s)
- Vera A Martynyuk
- Institute of Cytology of Russian Academy of Sciences, ikhoretsky 4, Saint Petersburg 194064, Russian Federation
| | - Svetlana S Efimova
- Institute of Cytology of Russian Academy of Sciences, ikhoretsky 4, Saint Petersburg 194064, Russian Federation
| | - Anna I Malykhina
- Institute of Cytology of Russian Academy of Sciences, ikhoretsky 4, Saint Petersburg 194064, Russian Federation
| | - Olga S Ostroumova
- Institute of Cytology of Russian Academy of Sciences, ikhoretsky 4, Saint Petersburg 194064, Russian Federation.
| |
Collapse
|
3
|
Wang R, Li X, Xu Y, Li Y, Zhang W, Guo R, Song J. Progress, pharmacokinetics and future perspectives of luteolin modulating signaling pathways to exert anticancer effects: A review. Medicine (Baltimore) 2024; 103:e39398. [PMID: 39183411 PMCID: PMC11346905 DOI: 10.1097/md.0000000000039398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Luteolin (3, 4, 5, 7-tetrahydroxyflavone) are natural flavonoids widely found in vegetables, fruits and herbs, with anti-tumor, anti-inflammatory and antioxidant effects, and also play an anti-cancer effect in various cancers such as lung, breast, prostate, and liver cancer, etc. Specifically, the anti-cancer mechanism includes regulation of various signaling pathways to induce apoptosis of tumor cells, inhibition of tumor cell proliferation and metastasis, anti-angiogenesis, regulation of immune function, synergistic anti-cancer drugs and regulation of reactive oxygen species levels of tumor cells. Specific anti-cancer mechanisms include regulation of various signaling pathways to induce apoptosis, inhibition of tumor cell proliferation and metastasis, anti-angiogenesis, reversal of epithelial-mesenchymal transition, regulation of immune function, synergism with anti-cancer drugs and regulation of reactive oxygen species levels in tumor cells. This paper integrates the latest cutting-edge research on luteolin and combines it with the prospect of future clinical applications, aiming to explore the mechanism of luteolin exerting different anticancer effects through the regulation of different signaling pathways, so as to provide a practical theoretical basis for the use of luteolin in clinical treatment and hopefully provide some reference for the future research direction of luteolin.
Collapse
Affiliation(s)
- Rui Wang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
- Medical School of Nantong University, Nantong, PR China
| | - Xia Li
- Department of General Medicine, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Yanhan Xu
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Yangyang Li
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Weisong Zhang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Rongqi Guo
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| | - Jianxiang Song
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People’s Hospital, Yancheng, PR China
| |
Collapse
|
4
|
Rajendran P. Unveiling the power of flavonoids: A dynamic exploration of their impact on cancer through matrix metalloproteinases regulation. Biomedicine (Taipei) 2024; 14:12-28. [PMID: 38939095 PMCID: PMC11204124 DOI: 10.37796/2211-8039.1447] [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: 09/29/2023] [Revised: 11/15/2023] [Accepted: 12/22/2023] [Indexed: 06/29/2024] Open
Abstract
Cancer stands as a significant contributor to global mortality rates, primarily driven by its progression and widespread dissemination. Despite notable strides in cancer therapy, the efficacy of current treatment strategies is compromised due to their inherent toxicity and the emergence of chemoresistance. Consequently, there is a critical need to evaluate alternative therapeutic approaches, with natural compounds emerging as promising candidates, showcasing demonstrated anticancer capabilities in various research models. This review manuscript presents a comprehensive examination of the regulatory mechanisms governing the expression of matrix metalloproteinases (MMPs) and delves into the potential therapeutic role of flavonoids as agents exhibiting specific anticancer activity against MMPs. The primary aim of this study is to elucidate the diverse functions associated with MMP production in cancer and to investigate the potential of flavonoids in modulating MMP expression to inhibit metastasis.
Collapse
Affiliation(s)
- Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, Tamil Nadu, India
| |
Collapse
|
5
|
Lu HY, Mi FL, Chou CM, Lin C, Chen YY, Chu CY, Liu CY, Lee YLA, Shih CC, Cheng CH. Layer-by-layer assembly of quercetin-loaded zein/γPGA/low-molecular-weight chitosan/fucoidan nanosystem for targeting inflamed blood vessels. Int J Biol Macromol 2024; 267:131369. [PMID: 38580026 DOI: 10.1016/j.ijbiomac.2024.131369] [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: 12/26/2023] [Revised: 03/03/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Chitosan acts as a versatile carrier in polymeric nanoparticle (NP) for diverse drug administration routes. Delivery of antioxidants, such as quercetin (Qu) showcases potent antioxidant and anti-inflammatory properties for reduction of various cardiovascular diseases, but low water solubility limits uptake. To address this, we developed a novel layer-by-layer zein/gamma-polyglutamic acid (γPGA)/low-molecular-weight chitosan (LC)/fucoidan NP for encapsulating Qu and targeting inflamed vessel endothelial cells. We used zein (Z) and γPGA (r) to encapsulate Qu (Qu-Zr NP) exhibited notably higher encapsulation efficiency compared to zein alone. Qu-Zr NP coated with LC (Qu-ZrLC2 NP) shows a lower particle size (193.2 ± 2.9 nm), and a higher zeta potential value (35.2 ± 0.4 mV) by zeta potential and transmission electron microscopy analysis. After coating Qu-ZrLC2 NP with fucoidan, Qu-ZrLC2Fa NP presented particle size (225.16 ± 0.92 nm), zeta potential (-25.66 ± 0.51 mV) and maintained antioxidant activity. Further analysis revealed that Qu-ZrLC2Fa NP were targeted and taken up by HUVEC cells and EA.hy926 endothelial cells. Notably, we observed Qu-ZrLC2Fa NP targeting zebrafish vessels and isoproterenol-induced inflamed vessels of rat. Our layer-by-layer formulated zein/γPGA/LC/fucoidan NP show promise as a targeted delivery system for water-insoluble drugs. Qu-ZrLC2Fa NP exhibit potential as an anti-inflammatory therapeutic for blood vessels.
Collapse
Affiliation(s)
- Hsin-Ying Lu
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan; Department of Physical Medicine and Rehabilitation, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Fwu-Long Mi
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Ming Chou
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chi Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Yu Chen
- School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Cheng-Ying Chu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan; CRISPR Gene Targeting Core Lab, Taipei Medical University, Taipei 11031, Taiwan
| | - Cheng-Yang Liu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Lin Amy Lee
- Departments of Medicine and Pediatrics, Hospice and Palliative Medicine, Duke University Hospital, Durham, NC 27710, USA
| | - Chun Che Shih
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan; Department of Physical Medicine and Rehabilitation, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
6
|
Fakhri S, Moradi SZ, Faraji F, Kooshki L, Webber K, Bishayee A. Modulation of hypoxia-inducible factor-1 signaling pathways in cancer angiogenesis, invasion, and metastasis by natural compounds: a comprehensive and critical review. Cancer Metastasis Rev 2024; 43:501-574. [PMID: 37792223 DOI: 10.1007/s10555-023-10136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Tumor cells employ multiple signaling mediators to escape the hypoxic condition and trigger angiogenesis and metastasis. As a critical orchestrate of tumorigenic conditions, hypoxia-inducible factor-1 (HIF-1) is responsible for stimulating several target genes and dysregulated pathways in tumor invasion and migration. Therefore, targeting HIF-1 pathway and cross-talked mediators seems to be a novel strategy in cancer prevention and treatment. In recent decades, tremendous efforts have been made to develop multi-targeted therapies to modulate several dysregulated pathways in cancer angiogenesis, invasion, and metastasis. In this line, natural compounds have shown a bright future in combating angiogenic and metastatic conditions. Among the natural secondary metabolites, we have evaluated the critical potential of phenolic compounds, terpenes/terpenoids, alkaloids, sulfur compounds, marine- and microbe-derived agents in the attenuation of HIF-1, and interconnected pathways in fighting tumor-associated angiogenesis and invasion. This is the first comprehensive review on natural constituents as potential regulators of HIF-1 and interconnected pathways against cancer angiogenesis and metastasis. This review aims to reshape the previous strategies in cancer prevention and treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6714415153, Iran
| | - Kassidy Webber
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA.
| |
Collapse
|
7
|
Shi M, Chen Z, Gong H, Peng Z, Sun Q, Luo K, Wu B, Wen C, Lin W. Luteolin, a flavone ingredient: Anticancer mechanisms, combined medication strategy, pharmacokinetics, clinical trials, and pharmaceutical researches. Phytother Res 2024; 38:880-911. [PMID: 38088265 DOI: 10.1002/ptr.8066] [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: 08/16/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 02/15/2024]
Abstract
Current pharmaceutical research is energetically excavating the pharmacotherapeutic role of herb-derived ingredients in multiple malignancies' targeting. Luteolin is one of the major phytochemical components that exist in various traditional Chinese medicine or medical herbs. Mounting evidence reveals that this phytoconstituent endows prominent therapeutic actions on diverse malignancies, with the underlying mechanisms, combined medication strategy, and pharmacokinetics elusive. Additionally, the clinical trial and pharmaceutical investigation of luteolin remain to be systematically delineated. The present review aimed to comprehensively summarize the updated information with regard to the anticancer mechanism, combined medication strategies, pharmacokinetics, clinical trials, and pharmaceutical researches of luteolin. The survey corroborates that luteolin executes multiple anticancer effects mainly by dampening proliferation and invasion, spurring apoptosis, intercepting cell cycle, regulating autophagy and immune, inhibiting inflammatory response, inducing ferroptosis, and pyroptosis, as well as epigenetic modification, and so on. Luteolin can be applied in combination with numerous clinical anticarcinogens and natural ingredients to synergistically enhance the therapeutic efficacy of malignancies while reducing adverse reactions. For pharmacokinetics, luteolin has an unfavorable oral bioavailability, it mainly persists in plasma as glucuronides and sulfate-conjugates after being metabolized, and is regarded as potent inhibitors of OATP1B1 and OATP2B1, which may be messed with the pharmacokinetic interactions of miscellaneous bioactive substances in vivo. Besides, pharmaceutical innovation of luteolin with leading-edge drug delivery systems such as host-guest complexes, nanoparticles, liposomes, nanoemulsion, microspheres, and hydrogels are beneficial to the exploitation of luteolin-based products. Moreover, some registered clinical trials on luteolin are being carried out, yet clinical research on anticancer effects should be continuously promoted.
Collapse
Affiliation(s)
- Mingyi Shi
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zixian Chen
- College of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Gong
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhaolei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiang Sun
- Sichuan Provincial Key Laboratory of Individualized Drug Therapy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Kaipei Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Baoyu Wu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuanbiao Wen
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Lin
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Raina R, Hussain A, Almutary AG, Haque S, Raza T, D’Souza AC, Subramani S, Sajeevan A. Co-administration of Chrysin and Luteolin with Cisplatin and Topotecan Exhibits a Variable Therapeutic Value in Human Cancer Cells, HeLa. ACS OMEGA 2023; 8:41204-41213. [PMID: 37970041 PMCID: PMC10633856 DOI: 10.1021/acsomega.3c04443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 11/17/2023]
Abstract
Combinational treatment is a promising strategy for better cancer treatment outcomes. Chrysin and luteolin have demonstrated effective anticancer activity. Cisplatin and topotecan are commonly used for the treatment of human cancers. However, various side effects including drug resistance are an imperative restriction to use them as pharmacological therapy. Therefore, the aim was to use these agents in combination with flavones for better efficacy. In the present study, it was found that the combination of chrysin and cisplatin and luteolin and cisplatin significantly improved the anticancer effect as both the combinations showed synergistic interactions [combinational index (CI < 1)]. Remarkably, the combination of chrysin and luteolin with topotecan depicted the antagonistic interaction (CI > 1). Further, increased expression of the pro-apoptotic proteins Bax and caspase 8 and the inhibition of the antiapoptotic protein Bcl-2 were instituted in the synergistic doses (chrysin + cisplatin and luteolin + cisplatin), hence promoting apoptosis. Also, it was found that the synergistic combination inhibited the migration of HeLa cells by downregulation of metalloproteases and upregulation of TIMPs. However, there are no significant changes depicted in the antagonistic combinations which support their role in their antagonistic effects. Based on these results, it can be inferred that the two or more drug combinations need to be explored well for their interaction to enhance the therapeutic outcomes.
Collapse
Affiliation(s)
- Ritu Raina
- School
of Life Sciences, Manipal Academy of Higher
of Education, Academic City 345050, Dubai, United Arab Emirates
| | - Arif Hussain
- School
of Life Sciences, Manipal Academy of Higher
of Education, Academic City 345050, Dubai, United Arab Emirates
| | - Abdulmajeed G. Almutary
- Department
of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Khalifa
City, Abu Dhabi 51072, United Arab Emirates
- Department
of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| | - Shafiul Haque
- Research
and Scientific Studies Unit, College of Nursing and Allied Health
Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Tasleem Raza
- Department
of Biochemistry, Era’s Lucknow Medical
College and Hospital, Lucknow 226003, India
| | - Ashley Cletus D’Souza
- School
of Life Sciences, Manipal Academy of Higher
of Education, Academic City 345050, Dubai, United Arab Emirates
| | - Sachin Subramani
- School
of Life Sciences, Manipal Academy of Higher
of Education, Academic City 345050, Dubai, United Arab Emirates
| | - Akash Sajeevan
- School
of Life Sciences, Manipal Academy of Higher
of Education, Academic City 345050, Dubai, United Arab Emirates
| |
Collapse
|
9
|
Hussain MS, Gupta G, Goyal A, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Fuloria S, Meenakshi DU, Jakhmola V, Pandey M, Singh SK, Dua K. From nature to therapy: Luteolin's potential as an immune system modulator in inflammatory disorders. J Biochem Mol Toxicol 2023; 37:e23482. [PMID: 37530602 DOI: 10.1002/jbt.23482] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023]
Abstract
Inflammation is an essential immune response that helps fight infections and heal tissues. However, chronic inflammation has been linked to several diseases, including cancer, autoimmune disorders, cardiovascular diseases, and neurological disorders. This has increased interest in finding natural substances that can modulate the immune system inflammatory signaling pathways to prevent or treat these diseases. Luteolin is a flavonoid found in many fruits, vegetables, and herbs. It has been shown to have anti-inflammatory effects by altering signaling pathways in immune cells. This review article discusses the current research on luteolin's role as a natural immune system modulator of inflammatory signaling mechanisms, such as its effects on nuclear factor-kappa B, mitogen-activated protein kinases, Janus kinase/signal transducer and activator of transcription, and inflammasome signaling processes. The safety profile of luteolin and its potential therapeutic uses in conditions linked to inflammation are also discussed. Overall, the data point to Luteolin's intriguing potential as a natural regulator of immune system inflammatory signaling processes. More research is needed to fully understand its mechanisms of action and possible therapeutic applications.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
- Center for Global Health research (CGHR), Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
| | | | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | | | | | - Vikas Jakhmola
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Manisha Pandey
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
10
|
Jomova K, Raptova R, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Valko M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: chronic diseases and aging. Arch Toxicol 2023; 97:2499-2574. [PMID: 37597078 PMCID: PMC10475008 DOI: 10.1007/s00204-023-03562-9] [Citation(s) in RCA: 276] [Impact Index Per Article: 276.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 07/24/2023] [Indexed: 08/21/2023]
Abstract
A physiological level of oxygen/nitrogen free radicals and non-radical reactive species (collectively known as ROS/RNS) is termed oxidative eustress or "good stress" and is characterized by low to mild levels of oxidants involved in the regulation of various biochemical transformations such as carboxylation, hydroxylation, peroxidation, or modulation of signal transduction pathways such as Nuclear factor-κB (NF-κB), Mitogen-activated protein kinase (MAPK) cascade, phosphoinositide-3-kinase, nuclear factor erythroid 2-related factor 2 (Nrf2) and other processes. Increased levels of ROS/RNS, generated from both endogenous (mitochondria, NADPH oxidases) and/or exogenous sources (radiation, certain drugs, foods, cigarette smoking, pollution) result in a harmful condition termed oxidative stress ("bad stress"). Although it is widely accepted, that many chronic diseases are multifactorial in origin, they share oxidative stress as a common denominator. Here we review the importance of oxidative stress and the mechanisms through which oxidative stress contributes to the pathological states of an organism. Attention is focused on the chemistry of ROS and RNS (e.g. superoxide radical, hydrogen peroxide, hydroxyl radicals, peroxyl radicals, nitric oxide, peroxynitrite), and their role in oxidative damage of DNA, proteins, and membrane lipids. Quantitative and qualitative assessment of oxidative stress biomarkers is also discussed. Oxidative stress contributes to the pathology of cancer, cardiovascular diseases, diabetes, neurological disorders (Alzheimer's and Parkinson's diseases, Down syndrome), psychiatric diseases (depression, schizophrenia, bipolar disorder), renal disease, lung disease (chronic pulmonary obstruction, lung cancer), and aging. The concerted action of antioxidants to ameliorate the harmful effect of oxidative stress is achieved by antioxidant enzymes (Superoxide dismutases-SODs, catalase, glutathione peroxidase-GPx), and small molecular weight antioxidants (vitamins C and E, flavonoids, carotenoids, melatonin, ergothioneine, and others). Perhaps one of the most effective low molecular weight antioxidants is vitamin E, the first line of defense against the peroxidation of lipids. A promising approach appears to be the use of certain antioxidants (e.g. flavonoids), showing weak prooxidant properties that may boost cellular antioxidant systems and thus act as preventive anticancer agents. Redox metal-based enzyme mimetic compounds as potential pharmaceutical interventions and sirtuins as promising therapeutic targets for age-related diseases and anti-aging strategies are discussed.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Renata Raptova
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, 812 37, Slovakia
| | - Suliman Y Alomar
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Saleh H Alwasel
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, 812 37, Slovakia.
| |
Collapse
|
11
|
Baloghová J, Michalková R, Baranová Z, Mojžišová G, Fedáková Z, Mojžiš J. Spice-Derived Phenolic Compounds: Potential for Skin Cancer Prevention and Therapy. Molecules 2023; 28:6251. [PMID: 37687080 PMCID: PMC10489044 DOI: 10.3390/molecules28176251] [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: 07/29/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Skin cancer is a condition characterized by the abnormal growth of skin cells, primarily caused by exposure to ultraviolet (UV) radiation from the sun or artificial sources like tanning beds. Different types of skin cancer include melanoma, basal cell carcinoma, and squamous cell carcinoma. Despite the advancements in targeted therapies, there is still a need for a safer, highly efficient approach to preventing and treating cutaneous malignancies. Spices have a rich history dating back thousands of years and are renowned for their ability to enhance the flavor, taste, and color of food. Derived from various plant parts like seeds, fruits, bark, roots, or flowers, spices are important culinary ingredients. However, their value extends beyond the culinary realm. Some spices contain bioactive compounds, including phenolic compounds, which are known for their significant biological effects. These compounds have attracted attention in scientific research due to their potential health benefits, including their possible role in disease prevention and treatment, such as cancer. This review focuses on examining the potential of spice-derived phenolic compounds as preventive or therapeutic agents for managing skin cancers. By compiling and analyzing the available knowledge, this review aims to provide insights that can guide future research in identifying new anticancer phytochemicals and uncovering additional mechanisms for combating skin cancer.
Collapse
Affiliation(s)
- Janette Baloghová
- Department of Dermatovenerology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (J.B.); (Z.B.); (Z.F.)
| | - Radka Michalková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia;
| | - Zuzana Baranová
- Department of Dermatovenerology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (J.B.); (Z.B.); (Z.F.)
| | - Gabriela Mojžišová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia;
| | - Zuzana Fedáková
- Department of Dermatovenerology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (J.B.); (Z.B.); (Z.F.)
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Kosice, Slovakia;
| |
Collapse
|
12
|
Gopinatha Pillai MS, Aiswarya SU, Keerthana CK, Rayginia TP, Anto RJ. Targeting receptor tyrosine kinase signaling: Avenues in the management of cutaneous squamous cell carcinoma. iScience 2023; 26:106816. [PMID: 37235052 PMCID: PMC10206193 DOI: 10.1016/j.isci.2023.106816] [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] [Indexed: 05/28/2023] Open
Abstract
Non-melanoma skin cancer (NMSC) is the most frequently diagnosed cancer worldwide. Among the various types of NMSCs, cutaneous squamous cell carcinoma (cSCC) exhibits more aggressive phenotype and is also the second-most prevalent type. Receptor tyrosine kinases (RTK) triggers key signaling events that play critical roles in the development of various cancers including cSCC. Unsurprisingly, for this reason, this family of proteins has become the cynosure of anti-cancer drug discovery pipelines and is also being considered as attractive targets against cSCC. Though inhibition of RTKs in cSCC has yielded favourable results, there is still scope for bettering the therapeutic outcome. In this review, we discuss the relevance of RTK signaling in the progression of cutaneous squamous cell carcinoma, and observations from clinical trials that used RTK inhibitors against cSCC. Backed by results from preclinical studies, including those from our lab, we also give insights into the scope of using some natural products as effective suppressors of RTK signaling and skin carcinogenesis.
Collapse
Affiliation(s)
| | - Sreekumar U. Aiswarya
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Chenicheri K. Keerthana
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Tennyson P. Rayginia
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
13
|
Rocchetti MT, Bellanti F, Zadorozhna M, Fiocco D, Mangieri D. Multi-Faceted Role of Luteolin in Cancer Metastasis: EMT, Angiogenesis, ECM Degradation and Apoptosis. Int J Mol Sci 2023; 24:ijms24108824. [PMID: 37240168 DOI: 10.3390/ijms24108824] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/09/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Luteolin (3',4',5,7-tetrahydroxyflavone), a member of the flavonoid family derived from plants and fruits, shows a wide range of biomedical applications. In fact, due to its anti-inflammatory, antioxidant and immunomodulatory activities, Asian medicine has been using luteolin for centuries to treat several human diseases, including arthritis, rheumatism, hypertension, neurodegenerative disorders and various infections. Of note, luteolin displays many anti-cancer/anti-metastatic properties. Thus, the purpose of this review consists in highlighting the relevant mechanisms by which luteolin inhibits tumor progression in metastasis, i.e., affecting epithelial-mesenchymal transition (EMT), repressing angiogenesis and lysis of extracellular matrix (ECM), as well as inducing apoptosis.
Collapse
Affiliation(s)
- Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| | - Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| | - Mariia Zadorozhna
- Medical Genetics Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 14, 27100 Pavia, Italy
| | - Daniela Fiocco
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| |
Collapse
|
14
|
Imran M, Insaf A, Hasan N, Sugandhi VV, Shrestha D, Paudel KR, Jha SK, Hansbro PM, Dua K, Devkota HP, Mohammed Y. Exploring the Remarkable Chemotherapeutic Potential of Polyphenolic Antioxidants in Battling Various Forms of Cancer. Molecules 2023; 28:molecules28083475. [PMID: 37110709 PMCID: PMC10142939 DOI: 10.3390/molecules28083475] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Plant-derived compounds, specifically antioxidants, have played an important role in scavenging the free radicals present under diseased conditions. The persistent generation of free radicals in the body leads to inflammation and can result in even more severe diseases such as cancer. Notably, the antioxidant potential of various plant-derived compounds prevents and deregulates the formation of radicals by initiating their decomposition. There is a vast literature demonstrating antioxidant compounds' anti-inflammatory, anti-diabetic, and anti-cancer potential. This review describes the molecular mechanism of various flavonoids, such as quercetin, kaempferol, naringenin, epicatechin, and epicatechin gallate, against different cancers. Additionally, the pharmaceutical application of these flavonoids against different cancers using nanotechnologies such as polymeric, lipid-based nanoparticles (solid-lipid and liquid-lipid), liposomes, and metallic nanocarriers is addressed. Finally, combination therapies in which these flavonoids are employed along with other anti-cancer agents are described, indicating the effective therapies for the management of various malignancies.
Collapse
Affiliation(s)
- Mohammad Imran
- Therapeutics Research Group, Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Areeba Insaf
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Vrushabh V Sugandhi
- Department of Pharmaceutics, Y.B. Chavan College of Pharmacy, Aurangabad 431001, India
| | - Deumaya Shrestha
- Department of Bioscience, Mokp o National University, Muna 58554, Republic of Korea
| | - Keshav Raj Paudel
- Centre of Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Saurav Kumar Jha
- Department of Biomedicine, Health & Life Convergence Sciences, Mokpo National University, Muna 58554, Republic of Korea
| | - Philip M Hansbro
- Centre of Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - 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, NSW 2007, Australia
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
- Pharmacy Program, Gandaki University, Pokhara 33700, Nepal
| | - Yousuf Mohammed
- Therapeutics Research Group, Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
15
|
Čižmárová B, Hubková B, Tomečková V, Birková A. Flavonoids as Promising Natural Compounds in the Prevention and Treatment of Selected Skin Diseases. Int J Mol Sci 2023; 24:ijms24076324. [PMID: 37047297 PMCID: PMC10094312 DOI: 10.3390/ijms24076324] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/22/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
Phytochemicals represent a large and diverse group of naturally occurring compounds, bioactive nutrients, or phytonutrients produced by plants, widely found in fruits, vegetables, whole grains products, legumes, beans, herbs, seeds, nuts, tea, and dark chocolate. They are classified according to their chemical structures and functional properties. Flavonoids belong to the phenolic class of phytochemicals with potential solid pharmacological effects as modulators of multiple signal transduction pathways. Their beneficial effect on the human body is associated with their antioxidant, anti-inflammatory, antimutagenic, and anticarcinogenic properties. Flavonoids are also widely used in various nutritional, pharmaceutical, medical, and cosmetic applications. In our review, we discuss the positive effect of flavonoids on chronic skin diseases such as vitiligo, psoriasis, acne, and atopic dermatitis.
Collapse
|
16
|
Costa AR, Duarte AC, Costa-Brito AR, Gonçalves I, Santos CRA. Bitter taste signaling in cancer. Life Sci 2023; 315:121363. [PMID: 36610638 DOI: 10.1016/j.lfs.2022.121363] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023]
Abstract
Pharmacoresistance of cancer cells to many drugs used in chemotherapy remains a major challenge for the treatment of cancer. Multidrug resistance transporters, especially ATP-binding cassette (ABC) transporters, are a major cause of cancer drug resistance since they translocate a broad range of drug compounds across the cell membrane, extruding them out of the cells. The regulation of ABC transporters by bitter taste receptors (TAS2Rs), which might be activated by specific bitter tasting compounds, was described in several types of cells/organs, becoming a potential target for cancer therapy. TAS2Rs expression has been reported in many organs and several types of cancer, like breast, ovarian, prostate, and colorectal cancers, where their activation was shown to be involved in various biological actions (cell survival, apoptosis, molecular transport, among others). Moreover, many TAS2Rs' ligands, such as flavonoids and alkaloids, with well-recognized beneficial properties, including several anticancer effects, have been reported as potential adjuvants in cancer therapies. In this review, we discuss the potential therapeutic role of TAS2Rs and bitter tasting compounds in different types of cancer as a possible way to circumvent chemoresistance.
Collapse
Affiliation(s)
- Ana R Costa
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal; CPIRN-IPG - Centro de Potencial e Inovação de Recursos Naturais, Instituto Politécnico da Guarda, Guarda, Portugal
| | - Ana R Costa-Brito
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal; Research Unit for Inland Development (UDI), Polytechnic of Guarda, Guarda, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal.
| |
Collapse
|
17
|
Wang Z, Liu F, Huang C, Zhang J, Wu J. Bufalin inhibits epithelial-mesenchymal transition and increases radiosensitivity of non-small cell lung cancer via inhibition of the Src signaling. J Thorac Dis 2023; 15:123-134. [PMID: 36794138 PMCID: PMC9922603 DOI: 10.21037/jtd-22-1859] [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: 11/11/2022] [Accepted: 01/10/2023] [Indexed: 01/17/2023]
Abstract
Background Epithelial-mesenchymal transition (EMT) is a biological process involved in tumor migration, invasion, and radiotherapy resistance. Bufalin can affect the proliferation, apoptosis and invasion of tumor cells by regulating multiple signaling pathways. Whether bufalin can increase radiosensitivity through EMT deserves further investigation. Methods In this study, we investigated the effect of bufalin on the EMT and radiosensitivity of non-small cell lung cancer (NSCLC) and the underlying molecular mechanism. NSCLC cells were treated with bufalin (at a dose of 0-100 nM) or irradiated with 6 MV X-rays (4 Gy/min). The effects of bufalin on cell survival, cell cycle, radiosensitivity, cell migration, and invasion were detected. Western blot was used to analyze the gene expression changes of Src signaling in NSCLC cell induced by Bufalin. Results Bufalin significantly inhibited cell survival, migration, and invasion and induced G2/M arrest and apoptosis. Cells co-treated with bufalin and radiation manifested a higher inhibitory effect compared to those treated with radiation or bufalin alone. Furthermore, the levels of p-Src and p-STAT3 were considerably reduced following bufalin treatment. Interestingly, elevated p-Src and p-STAT3 were observed in cells treated with radiation. Bufalin inhibited radiation-induced p-Src and p-STAT3, whereas the knockdown of Src abrogated the effects of bufalin on cell migration, invasion, EMT, and radiosensitivity. Conclusions Bufalin inhibits EMT and enhances radiosensitivity through targeting Src signaling in NSCLC.
Collapse
Affiliation(s)
- Zijian Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fen Liu
- Cancer Institute, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Chen Huang
- Cancer Institute, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Jiaqi Zhang
- Cancer Institute, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
18
|
Wen SY, Wei BY, Ma JQ, Wang L, Chen YY. Phytochemicals, Biological Activities, Molecular Mechanisms, and Future Prospects of Plantago asiatica L. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:143-173. [PMID: 36545763 DOI: 10.1021/acs.jafc.2c07735] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Plantago asiatica L. has been used as a vegetable and nutritious food in Asia for thousands of years. According to recent phytochemical and pharmacological research, the active compositions of the plant contribute to various health benefits, such as antioxidant, anti-inflammatory, antibacterial, antiviral, and anticancer. This article reviews the 87 components of the plant and their structures, as well as their biological activities and molecular research progress, in detail. This review provides valuable reference material for further study, production, and application of P. asiatica, as well as its components in functional foods and therapeutic agents.
Collapse
Affiliation(s)
- Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Bing-Yan Wei
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Jie-Qiong Ma
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Li Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Yan-Yan Chen
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
19
|
Investigation of the Active Compounds and Important Pathways of Huaiqihuang Granule for the Treatment of Immune Thrombocytopenia Using Network Pharmacology and Molecular Docking. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5984361. [PMID: 36660453 PMCID: PMC9845056 DOI: 10.1155/2023/5984361] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/20/2022] [Accepted: 12/26/2022] [Indexed: 01/11/2023]
Abstract
Materials and Methods Compounds of HQHG were scanned by LC-MS/MS, and the target profiles of compounds were identified based on SwissTarget Prediction. ITP target proteins were collected from various databases. Then, KEGG pathway and GO enrichment analyses were performed to explore the signaling pathways related to HQHG for ITP. The PPI and drug-herbs-compounds-targets-pathways network were constructed using Cytoscape 3.7.2. Finally, Discovery studio software was used to confirm the key targets and active compounds from HQHG. Results A total of 187 interacting targets of 19 potentially active compounds in HQHG and 3837 ITP-related targets were collected. Then, 187 intersection targets were obtained. A total of 20 key targets including EGFR, CASP3, TNF, STAT3, and ERBB2 were identified through PPI network analysis. These targets were mainly focused on the biological processes of positive regulation of protein phosphorylation, cellular response to organonitrogen compound, and cellular response to nitrogen compound. 20 possible pathways of HQHG in the treatment of ITP were identified through KEGG enrichment. EGFR, CASP3, TNF, and STAT3 are the four most important target proteins, while adenosine, caffeic acid, ferulic acid, quercetin-3β-D-glucoside, rutin, scopoletin, and tianshic acid are the most important active compounds, which were validated by molecular docking simulation. Conclusion This study demonstrated that HQHG produced relief effects against ITP by regulating multitargets and multipathways with multicompounds. And the combined data provide novel insight of drug developing for ITP.
Collapse
|
20
|
A low-molecular-weight chitosan fluorometric-based assay for evaluating antiangiogenic drugs. Int J Biol Macromol 2022; 224:927-937. [DOI: 10.1016/j.ijbiomac.2022.10.178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
|
21
|
Talib WH, Awajan D, Hamed RA, Azzam AO, Mahmod AI, AL-Yasari IH. Combination Anticancer Therapies Using Selected Phytochemicals. Molecules 2022; 27:5452. [PMID: 36080219 PMCID: PMC9458090 DOI: 10.3390/molecules27175452] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is still one of the most widespread diseases globally, it is considered a vital health challenge worldwide and one of the main barriers to long life expectancy. Due to the potential toxicity and lack of selectivity of conventional chemotherapeutic agents, discovering alternative treatments is a top priority. Plant-derived natural products have high potential in cancer treatment due to their multiple mechanisms of action, diversity in structure, availability in nature, and relatively low toxicity. In this review, the anticancer mechanisms of the most common phytochemicals were analyzed. Furthermore, a detailed discussion of the anticancer effect of combinations consisting of natural product or natural products with chemotherapeutic drugs was provided. This review should provide a strong platform for researchers and clinicians to improve basic and clinical research in the development of alternative anticancer medicines.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Dima Awajan
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Reem Ali Hamed
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Aya O. Azzam
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Intisar Hadi AL-Yasari
- Department of Genetic Engineering, College of Biotechnology, Al-Qasim Green University, Babylon 964, Iraq
| |
Collapse
|
22
|
Ahsan H, Islam SU, Ahmed MB, Lee YS. Role of Nrf2, STAT3, and Src as Molecular Targets for Cancer Chemoprevention. Pharmaceutics 2022; 14:1775. [PMID: 36145523 PMCID: PMC9505731 DOI: 10.3390/pharmaceutics14091775] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/23/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is a complex and multistage disease that affects various intracellular pathways, leading to rapid cell proliferation, angiogenesis, cell motility, and migration, supported by antiapoptotic mechanisms. Chemoprevention is a new strategy to counteract cancer; to either prevent its incidence or suppress its progression. In this strategy, chemopreventive agents target molecules involved in multiple pathways of cancer initiation and progression. Nrf2, STAT3, and Src are promising molecular candidates that could be targeted for chemoprevention. Nrf2 is involved in the expression of antioxidant and phase II metabolizing enzymes, which have direct antiproliferative action as well as indirect activities of reducing oxidative stress and eliminating carcinogens. Similarly, its cross-talk with NF-κB has great anti-inflammatory potential, which can be utilized in inflammation-induced/associated cancers. STAT3, on the other hand, is involved in multiple pathways of cancer initiation and progression. Activation, phosphorylation, dimerization, and nuclear translocation are associated with tumor cell proliferation and angiogenesis. Src, being the first oncogene to be discovered, is important due to its convergence with many upstream stimuli, its cross-talk with other potential molecular targets, such as STAT3, and its ability to modify the cell cytoskeleton, making it important in cancer invasion and metastasis. Therefore, the development of natural/synthetic molecules and/or design of a regimen that can reduce oxidative stress and inflammation in the tumor microenvironment and stop multiple cellular targets in cancer to stop its initiation or retard its progression can form newer chemopreventive agents.
Collapse
Affiliation(s)
- Haseeb Ahsan
- Department of Pharmacy, Faculty of Life and Environmental Sciences, University of Peshawar, Peshawar 25120, Pakistan
| | - Salman Ul Islam
- Department of Pharmacy, CECOS University, Peshawar 25000, Pakistan
| | - Muhammad Bilal Ahmed
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Young Sup Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
23
|
Cirsilineol Inhibits the Proliferation of Human Prostate Cancer Cells by Inducing Reactive Oxygen Species (ROS)-Mediated Apoptosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7975664. [PMID: 35855832 PMCID: PMC9288295 DOI: 10.1155/2022/7975664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 11/18/2022]
Abstract
Cirsilineol has been reported to exhibit anticancer effects against several human cancer cell lines. The present study was designed to evaluate the anticancer effects of cirsilineol against the human DU-145 prostate cancer cells. The results showed that cirsilineol suppressed the proliferation of DU-145 cancer cells in a dose-dependent manner with minimal cytotoxic effects against the normal cells. The IC50 of cirsilineol was found to be 7 μM and 110 μM against prostate cancer DU-145 and normal HPrEC prostate cells, respectively. Acridine orange and ethidium bromide (AO/EB) staining showed that cirsilineol induced apoptosis in DU-145 prostate cancer cells. The Annexin V/PI staining further confirmed the induction of apoptosis in DU-145 cells. The western blot analysis showed that cirsilineol suppressed the expression of Bax and upregulated the expression of Bcl-2 in prostate cancer DU-145 cells. Moreover, cirsilineol caused a dose-dependent increase in reactive oxygen species (ROS) levels in prostate cancer. Wound healing and Transwell assays showed that cirsilineol inhibits migration and invasion of DU-145 prostate cancer cells. Summing up, the results suggest that cirsilineol suppresses the proliferation of prostate cancer cells and may prove to be a beneficial lead molecule for the development of chemotherapy for prostate cancer.
Collapse
|
24
|
Eltamany EE, Goda MS, Nafie MS, Abu-Elsaoud AM, Hareeri RH, Aldurdunji MM, Elhady SS, Badr JM, Eltahawy NA. Comparative Assessment of the Antioxidant and Anticancer Activities of Plicosepalus acacia and Plicosepalus curviflorus: Metabolomic Profiling and In Silico Studies. Antioxidants (Basel) 2022; 11:antiox11071249. [PMID: 35883740 PMCID: PMC9311546 DOI: 10.3390/antiox11071249] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
This study presents a comparison between two mistletoe plants—P. acacia and P. curviflorus—regarding their total phenolic contents and antioxidant and anticancer activities. P. curviflorus exhibited a higher total phenolics content (340.62 ± 19.46 mg GAE/g extract), and demonstrated higher DPPH free radical scavenging activity (IC50 = 48.28 ± 3.41µg/mL), stronger reducing power (1.43 ± 0.54 mMol Fe+2/g) for ferric ions, and a greater total antioxidant capacity (41.89 ± 3.15 mg GAE/g) compared to P. acacia. The cytotoxic effects of P. acacia and P. curviflorus methanol extracts were examined on lung (A549), prostate (PC-3), ovarian (A2780) and breast (MDA-MB-231) cancer cells. The highest anticancer potential for the two extracts was observed on PC-3 prostate cancer cells, where P. curviflorus exhibited more pronounced antiproliferative activity (IC50 = 25.83 μg/mL) than P. acacia (IC50 = 34.12 μg/mL). In addition, both of the tested extracts arrested the cell cycle at the Pre-G1 and G1 phases, and induced apoptosis. However, P. curviflorus extract possessed the highest apoptotic effect, mediated by the upregulation of p53, Bax, and caspase-3, 8 and 9, and the downregulation of Bcl-2 expression. In the pursuit to link the chemical diversity of P. curviflorus with the exhibited bioactivities, its metabolomic profiling was achieved by the LC-ESI-TOF-MS/MS technique. This permitted the tentative identification of several phenolics—chiefly flavonoid derivatives, beside some triterpenes and sterols—in the P. curviflorus extract. Furthermore, all of the metabolites in P. curviflorus and P. acacia were inspected for their binding modes towards both CDK-2 and EGFR proteins using molecular docking studies in an attempt to understand the superiority of P. curviflorus over P. acacia regarding their antiproliferative effect on PC-3 cancer cells. Docking studies supported our experimental results; with all of this taken together, P. curviflorus could be regarded as a potential prospect for the development of chemotherapeutics for prostate cancer.
Collapse
Affiliation(s)
- Enas E. Eltamany
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.E.E.); (M.S.G.); (N.A.E.)
| | - Marwa S. Goda
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.E.E.); (M.S.G.); (N.A.E.)
| | - Mohamed S. Nafie
- Department of Chemistry, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt;
| | - Abdelghafar M. Abu-Elsaoud
- Department of Botany and Microbiology, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt;
| | - Rawan H. Hareeri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mohammed M. Aldurdunji
- Department of Clinical Pharmacy, College of Pharmacy, Umm Al-Qura University, P.O. Box 13578, Makkah 21955, Saudi Arabia;
| | - Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (S.S.E.); (J.M.B.); Tel.: +966-544512552 (S.S.E.); +20-1091332451 (J.M.B.)
| | - Jihan M. Badr
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.E.E.); (M.S.G.); (N.A.E.)
- Correspondence: (S.S.E.); (J.M.B.); Tel.: +966-544512552 (S.S.E.); +20-1091332451 (J.M.B.)
| | - Nermeen A. Eltahawy
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.E.E.); (M.S.G.); (N.A.E.)
| |
Collapse
|
25
|
Sadrkhanloo M, Entezari M, Orouei S, Ghollasi M, Fathi N, Rezaei S, Hejazi ES, Kakavand A, Saebfar H, Hashemi M, Goharrizi MASB, Salimimoghadam S, Rashidi M, Taheriazam A, Samarghandian S. STAT3-EMT axis in tumors: modulation of cancer metastasis, stemness and therapy response. Pharmacol Res 2022; 182:106311. [PMID: 35716914 DOI: 10.1016/j.phrs.2022.106311] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 02/07/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) mechanism is responsible for metastasis of tumor cells and their spread to various organs and tissues of body, providing undesirable prognosis. In addition to migration, EMT increases stemness and mediates therapy resistance. Hence, pathways involved in EMT regulation should be highlighted. STAT3 is an oncogenic pathway that can elevate growth rate and migratory ability of cancer cells and induce drug resistance. The inhibition of STAT3 signaling impairs cancer progression and promotes chemotherapy-mediated cell death. Present review focuses on STAT3 and EMT interaction in modulating cancer migration. First of all, STAT3 is an upstream mediator of EMT and is able to induce EMT-mediated metastasis in brain tumors, thoracic cancers and gastrointestinal cancers. Therefore, STAT3 inhibition significantly suppresses cancer metastasis and improves prognosis of patients. EMT regulators such as ZEB1/2 proteins, TGF-β, Twist, Snail and Slug are affected by STAT3 signaling to stimulate cancer migration and invasion. Different molecular pathways such as miRNAs, lncRNAs and circRNAs modulate STAT3/EMT axis. Furthermore, we discuss how STAT3 and EMT interaction affects therapy response of cancer cells. Finally, we demonstrate targeting STAT3/EMT axis by anti-tumor agents and clinical application of this axis for improving patient prognosis.
Collapse
Affiliation(s)
- Mehrdokht Sadrkhanloo
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sima Orouei
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Marzieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Nikoo Fathi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shamin Rezaei
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elahe Sadat Hejazi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirabbas Kakavand
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Saebfar
- European University Association, League of European Research Universities, University of Milan, Italy
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
26
|
Flavonoids as Potential Anti-Inflammatory Molecules: A Review. Molecules 2022; 27:molecules27092901. [PMID: 35566252 PMCID: PMC9100260 DOI: 10.3390/molecules27092901] [Citation(s) in RCA: 213] [Impact Index Per Article: 106.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022] Open
Abstract
Hydroxylated polyphenols, also called flavonoids, are richly present in vegetables, fruits, cereals, nuts, herbs, seeds, stems, and flowers of numerous plants. They possess numerous medicinal properties such as antioxidant, anti-cancer, anti-microbial, neuroprotective, and anti-inflammation. Studies show that flavonoids activate antioxidant pathways that render an anti-inflammatory effect. They inhibit the secretions of enzymes such as lysozymes and β-glucuronidase and inhibit the secretion of arachidonic acid, which reduces inflammatory reactions. Flavonoids such as quercetin, genistein, apigenin, kaempferol, and epigallocatechin 3-gallate modulate the expression and activation of a cytokine such as interleukin-1beta (IL-1β), Tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8); regulate the gene expression of many pro-inflammatory molecules such s nuclear factor kappa-light chain enhancer of activated B cells (NF-κB), activator protein-1 (AP-1), intercellular adhesion molecule-1 (ICAM), vascular cell adhesion molecule-1 (VCAM), and E-selectins; and also inhibits inducible nitric oxide (NO) synthase, cyclooxygenase-2, and lipoxygenase, which are pro-inflammatory enzymes. Understanding the anti-inflammatory action of flavonoids provides better treatment options, including coronavirus disease 2019 (COVID-19)-induced inflammation, inflammatory bowel disease, obstructive pulmonary disorder, arthritis, Alzheimer’s disease, cardiovascular disease, atherosclerosis, and cancer. This review highlights the sources, biochemical activities, and role of flavonoids in enhancing human health.
Collapse
|
27
|
Luteolin Causes 5'CpG Demethylation of the Promoters of TSGs and Modulates the Aberrant Histone Modifications, Restoring the Expression of TSGs in Human Cancer Cells. Int J Mol Sci 2022; 23:ijms23074067. [PMID: 35409426 PMCID: PMC8999529 DOI: 10.3390/ijms23074067] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer progression is linked to abnormal epigenetic alterations such as DNA methylation and histone modifications. Since epigenetic alterations, unlike genetic changes, are heritable and reversible, they have been considered as interesting targets for cancer prevention and therapy by dietary compounds such as luteolin. In this study, epigenetic modulatory behaviour of luteolin was analysed on HeLa cells. Various assays including colony forming and migration assays, followed by biochemical assays of epigenetic enzymes including DNA methyltransferase, histone methyl transferase, histone acetyl transferase, and histone deacetylases assays were performed. Furthermore, global DNA methylation and methylation-specific PCR for examining the methylation status of CpG promoters of various tumour suppressor genes (TSGs) and the expression of these TSGs at transcript and protein level were performed. It was observed that luteolin inhibited migration and colony formation in HeLa cells. It also modulated DNA methylation at promoters of TSGs and the enzymatic activity of DNMT, HDAC, HMT, and HAT and reduced the global DNA methylation. Decrease in methylation resulted in the reactivation of silenced tumour suppressor genes including FHIT, DAPK1, PTEN, CDH1, SOCS1, TIMPS, VHL, TP53, TP73, etc. Hence, luteolin-targeted epigenetic alterations provide a promising approach for cancer prevention and intervention.
Collapse
|
28
|
Juszczak AM, Wöelfle U, Končić MZ, Tomczyk M. Skin cancer, including related pathways and therapy and the role of luteolin derivatives as potential therapeutics. Med Res Rev 2022; 42:1423-1462. [PMID: 35187675 PMCID: PMC9303584 DOI: 10.1002/med.21880] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/16/2021] [Accepted: 01/23/2022] [Indexed: 12/12/2022]
Abstract
Cutaneous malignant melanoma is the fastest growing and the most aggressive form of skin cancer that is diagnosed. However, its incidence is relatively scarce compared to the highest mortality rate of all skin cancers. The much more common skin cancers include nonmelanoma malignant skin cancers. Moreover, over the past several decades, the frequency of all skin cancers has increased much more dynamically than that of almost any other type of cancer. Among the available therapeutic options for skin cancers, chemotherapy used immediately after the surgical intervention has been an essential element. Unfortunately, the main problem with conventional chemopreventive regimens involves the lack of response to treatment and the associated side effects. Hence, there is a need for much more effective anticancer drugs. Correspondingly, the targeted alternatives have involved phytochemicals, which are safer chemotherapeutic agents and exhibit competitive anticancer activity with high therapeutic efficacy. Among polyphenolic compounds, some flavonoids and their derivatives, which are mostly found in medicinal plants, have been demonstrated to influence the modulation of signaling pathways at each stage of the carcinogenesis process, which is also important in the context of skin cancers. Hence, this review focuses on an exhaustive overview of the therapeutic effects of luteolin and its derivatives in the treatment and prevention of skin cancers. The bioavailability and structure–activity relationships of luteolin derivatives are also discussed. This review is the first such complete account of all of the scientific reports concerning this particular group of natural compounds that target a specific area of neoplastic diseases.
Collapse
Affiliation(s)
- Aleksandra M. Juszczak
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine Medical University of Białystok Białystok Poland
| | - Ute Wöelfle
- Department of Dermatology and Venereology, Research Center Skinitial, Medical Center, Faculty of Medicine University of Freiburg Freiburg Germany
| | - Marijana Zovko Končić
- Department of Pharmacognosy, Faculty of Pharmacy and Biochemistry University of Zagreb Zagreb Croatia
| | - Michał Tomczyk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine Medical University of Białystok Białystok Poland
| |
Collapse
|
29
|
Slika H, Mansour H, Wehbe N, Nasser SA, Iratni R, Nasrallah G, Shaito A, Ghaddar T, Kobeissy F, Eid AH. Therapeutic potential of flavonoids in cancer: ROS-mediated mechanisms. Biomed Pharmacother 2022; 146:112442. [PMID: 35062053 DOI: 10.1016/j.biopha.2021.112442] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is a leading cause of morbidity and mortality around the globe. Reactive oxygen species (ROS) play contradicting roles in cancer incidence and progression. Antioxidants have attracted attention as emerging therapeutic agents. Among these are flavonoids, which are natural polyphenols with established anticancer and antioxidant capacities. Increasing evidence shows that flavonoids can inhibit carcinogenesis via suppressing ROS levels. Surprisingly, flavonoids can also trigger excessive oxidative stress, but this can also induce death of malignant cells. In this review, we explore the inherent characteristics that contribute to the antioxidant capacity of flavonoids, and we dissect the scenarios in which they play the contrasting role as pro-oxidants. Furthermore, we elaborate on the pathways that link flavonoid-mediated modulation of ROS to the prevention and treatment of cancer. Special attention is given to the ROS-mediated anticancer functions that (-)-epigallocatechin gallate (EGCG), hesperetin, naringenin, quercetin, luteolin, and apigenin evoke in various cancers. We also delve into the structure-function relations that make flavonoids potent antioxidants. This review provides a detailed perspective that can be utilized in future experiments or trials that aim at utilizing flavonoids or verifying their efficacy for developing new pharmacologic agents. We support the argument that flavonoids are attractive candidates for cancer therapy.
Collapse
Affiliation(s)
- Hasan Slika
- Department of Pharmacology and Toxicology, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon.
| | - Hadi Mansour
- Department of Pharmacology and Toxicology, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon.
| | - Nadine Wehbe
- Department of Biology, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon.
| | - Suzanne A Nasser
- Department of Pharmacology and Therapeutics, Beirut Arab University, P.O. Box 11-5020, Beirut, Lebanon.
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, P.O. Box 15551, Al-Ain, United Arab Emirates.
| | - Gheyath Nasrallah
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, P.O. Box: 2713, Doha, Qatar.
| | - Abdullah Shaito
- Biomedical Research Center, Qatar University, P.O. Box: 2713, Doha, Qatar.
| | - Tarek Ghaddar
- Department of Chemistry, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, P.O. Box: 11-0236, Beirut, Lebanon.
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
30
|
Lim HM, Park SH. Regulation of reactive oxygen species by phytochemicals for the management of cancer and diabetes. Crit Rev Food Sci Nutr 2022; 63:5911-5936. [PMID: 34996316 DOI: 10.1080/10408398.2022.2025574] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cancer and diabetes mellitus are served as typical life-threatening diseases with common risk factors. Developing therapeutic measures in cancers and diabetes have aroused attention for a long time. However, the problems with conventional treatments are in challenge, including side effects, economic burdens, and patient compliance. It is essential to secure safe and efficient therapeutic methods to overcome these issues. As an alternative method, antioxidant and pro-oxidant properties of phytochemicals from edible plants have come to the fore. Phytochemicals are naturally occurring compounds, considered promising agent applicable in treatment of various diseases with beneficial effects. Either antioxidative or pro-oxidative activity of various phytochemicals were found to contribute to regulation of cell proliferation, differentiation, cell cycle arrest, and apoptosis, which can exert preventive and therapeutic effects against cancer and diabetes. In this article, the antioxidant or pro-oxidant effects and underlying mechanisms of flavonoids, alkaloids, and saponins in cancer or diabetic models demonstrated by the recent studies are summarized.
Collapse
Affiliation(s)
- Heui Min Lim
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| |
Collapse
|
31
|
Recent advances in valorization of citrus fruits processing waste: a way forward towards environmental sustainability. Food Sci Biotechnol 2021; 30:1601-1626. [PMID: 34925937 DOI: 10.1007/s10068-021-00984-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Citrus fruits are well known for their medicinal and therapeutic potential due to the presence of immense bioactive components. With the enormous consumption of citrus juice, citrus processing industries are focused on the production of juice but at the same time, a large amount of waste is produced mainly in the form of peel, seeds, pomace, and wastewater. This waste left after processing leads to environmental pollution and health-related hazards. However, it could be exploited for the recovery of essential oils, pectin, nutraceuticals, macro and micronutrients, ethanol, and biofuel generation. In view of the importance and health benefits of bioactive compounds found in citrus waste, the present review summarizes the recent work done on the citrus fruit waste valorization for recovery of value-added compounds leading to zero wastage. Therefore, instead of calling it waste, these could be a good resource of significant valuable components, in this way encouraging the zero-waste theory.
Collapse
|
32
|
Ho JY, Lu HY, Cheng HH, Kuo YC, Lee YLA, Cheng CH. UBE2S activates NF-κB signaling by binding with IκBα and promotes metastasis of lung adenocarcinoma cells. Cell Oncol (Dordr) 2021; 44:1325-1338. [PMID: 34582005 DOI: 10.1007/s13402-021-00639-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/17/2021] [Indexed: 10/20/2022] Open
Abstract
PURPOSE Nuclear factor (NF)-κB signaling in cancer cells has been reported to be involved in tumorigenesis. Phosphorylation and degradation of inhibitor of NF-κBα (IκBα) is a canonical pathway of NF-κB signaling. Here, we aimed to identify and characterize noncanonical activation of NF-κB signaling by ubiquitin-conjugating enzyme E2S (UBE2S) in lung adenocarcinoma cells. METHODS TCGA and the Human Atlas Protein Database were used to analyze the survival rate of lung adenocarcinoma patients in conjunction with UBE2S expression. In addition, PC9, H460, H441 and A549 lung adenocarcinoma cells were used in this study. PC9 and H460 cells were selected for further analysis because they expressed different UBE2S protein levels. Specific IKK inhibitors, PS1145 and SC514, were used to assess IκBα phosphorylation. Western blot analysis was used to assess protein levels in PC9 and H460 cells. A scratch wound-healing assay was used to analyze the migrative abilities of PC9 and H460 cells. Overexpression and knockdown of UBE2S in H460 and PC9 cells were used to analyze their effects on downstream protein levels. Immunoprecipitation, immunofluorescent staining, glutathione S transferase (GST) pull-down and in vitro binding assays were used to analyze the interaction between UBE2S and IκBα. A luciferase assay was used to analyze activation of NF-κB signaling regulated by UBE2S. An in vivo zebrafish xenograft model was used to assess metastasis of PC9 cells regulated by UBE2S. RESULTS We found that UBE2S expression in lung adenocarcinoma patients was negatively related to survival rate. The protein level of UBE2S was higher in PC9 cells than in H460 cells, which was opposite to that observed for IκBα. PC9 cells showed a higher UBE2S expression and migrative ability than H460 cells. Phosphorylation of IκBα was not changed by treatment with the IKK-specific inhibitors PS1145 and SC514 in PC9 and H460 cells. Overexpression and knockdown of UBE2S in H460 and PC9 cells revealed that the protein levels of IκBα were inversely regulated. Immunoprecipitation, immunofluorescent staining, GST pull-down and in vitro binding assays revealed direct binding of UBE2S with IκBα. Nuclear P65 protein levels and luciferase assays showed that NF-κB signaling was regulated by UBE2S. The expression of epithelial-to-mesenchymal (EMT) markers and the migrative ability of lung adenocarcinoma cells were also regulated by UBE2S. A zebrafish xenograft tumor model showed a reduction in the metastasis of PC9 cells that was induced by UBE2S knockdown. CONCLUSIONS Higher UBE2S expression in lung adenocarcinomas may lead to increased binding with IκBα to activate NF-κB signaling and promote adenocarcinoma cell metastasis. UBE2S may serve as a potential therapeutic target for lung adenocarcinomas.
Collapse
Affiliation(s)
- Jhih-Yun Ho
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 11031, Taipei, Taiwan
| | - Hsin-Ying Lu
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, 11031, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, Wan Fang Hospital, Taipei Medical University, 11031, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, 11031, Taipei, Taiwan
| | - Hsing-Hsien Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 11031, Taipei, Taiwan
| | - Yu-Chieh Kuo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yu-Lin Amy Lee
- Departments of Medicine and Pediatrics, Duke University Hospital, Durham, NC, 27704, USA
| | - Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 11031, Taipei, Taiwan.
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, 11031, Taipei, Taiwan.
| |
Collapse
|
33
|
Balogun TA, Ige OM, Alausa AO, Onyeani CO, Tiamiyu ZA, Omoboyowa DA, Saibu OA, Abdullateef OT. Receptor tyrosine kinases as a therapeutic target by natural compounds in cancer treatment. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00346-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Receptor tyrosine kinases (RTKs) are single-pass transmembrane proteins that play significant roles in regulating cellular processes, including cell division and growth. Overexpression and mutations of RTKs have been found in clinical manifestations of different forms of cancer. Therefore, RTKs have received considerable interest as a therapeutic biomarker in the treatment of cancer cells.
Main body of the abstract
Comprehensive data on RTKs, pharmacological and biological properties of natural compounds were systematically searched up to 2021 using relevant keywords from various databases, such as Google Scholar, PubMed, Web of Science, and Scopus. The scientific search by various standard electronic resources and databases unveils the effectiveness of medicinal plants in the treatment of various cancers. In vitro and in vivo studies suggested that bioactive compounds such as flavonoids, phenols, alkaloids, and many others can be used pharmacologically as RTKs inhibitors (RTKI) either by competing with ATP at the ATP binding site of the tyrosine kinase domain or competing for the receptor extracellular domain. Additionally, studies conducted on animal models indicated that inhibition of RTKs catalytic activity by natural compounds is one of the most effective ways to block the activation of RTKs signaling cascades, thereby hampering the proliferation of cancer cells. Furthermore, various pharmacological experiments, transcriptomic, and proteomic data also reported that cancer cells treated with different plants extracts or isolated phytochemicals exhibited better anticancer properties with minimal side effects than synthetic drugs. Clinically, natural compounds have demonstrated significant anti-proliferative effect via induction of cell apoptosis in cancer cell lines.
Short conclusion
An in-depth knowledge of the mechanism of inhibition and structural characterization of RTKs is important to the design of novel and selective RTKIs. This review focuses on the molecular mechanisms and structures of natural compounds RTKI targeting vascular endothelial growth factor, epidermal growth factor receptor, insulin receptor, and platelet-derived growth factor while also giving future directions to ameliorate the scientific burden of cancer.
Graphic abstract
Collapse
|
34
|
Almeida TC, Seibert JB, Amparo TR, de Souza GHB, da Silva GN, Dos Santos DH. Modulation of Long Non-Coding RNAs by Different Classes of Secondary Metabolites from Plants: A Mini-Review on Antitumor Effects. Mini Rev Med Chem 2021; 22:1232-1255. [PMID: 34720079 DOI: 10.2174/1389557521666211101161548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/07/2021] [Accepted: 09/10/2021] [Indexed: 11/22/2022]
Abstract
The broad pharmacological spectrum of plants is related to their secondary metabolism, which is responsible for the synthesis of different compounds that have multiple effects on cellular physiology. Among the biological effects presented by phytochemicals, their use for the prevention and treatment of cancer can be highlighted. This occurs due to several mechanisms of antitumor action demonstrated by these compounds, including regulation of the cell signaling pathways and inhibition of tumor growth. In this way, long non-coding RNAs (lncRNAs) appear to be promising targets for the treatment of cancer. Their deregulation has already been related to a variety of clinical-pathological parameters. However, the effects of secondary metabolites on lncRNAs are still restricted. For this reason, the present review aimed to gather data on phytochemicals with action on lncRNAs in order to confirm their possible antitumor potential. According to the literature, terpenoid and flavonoid are the main examples of secondary metabolites involved with lncRNAs activity. In addition, the lncRNAs H19, CASC2, HOTAIR, NKILA, CCAT1, MALAT1, AFAP1-AS1, MEG3, and CDKN2B-AS1 can be highlighted as important targets in the search for new anti-tumor agents since they act as modulating pathways related to cell proliferation, cell cycle, apoptosis, cell migration and invasion. Finally, challenges for the use of natural products as a commercial drug were also discussed. The low yield, selectivity index and undesirable pharmacokinetic parameters were emphasized as a difficulty for obtaining these compounds on a large scale and for improving the potency of its biological effect. However, the synthesis and/or development of formulations were suggested as a possible approach to solve these problems. All of these data together confirm the potential of secondary metabolites as a source of new anti-tumor agents acting on lncRNAs.
Collapse
Affiliation(s)
- Tamires Cunha Almeida
- Department of Pharmacy, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto. Brazil
| | | | - Tatiane Roquete Amparo
- Department of Pharmacy, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto. Brazil
| | | | - Glenda Nicioli da Silva
- Department of Clinical Analysis, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto. Brazil
| | | |
Collapse
|
35
|
Wang T, Lyu CY, Jiang YH, Dong XY, Wang Y, Li ZH, Wang JX, Xu RR. A drug-biomarker interaction model to predict the key targets of Scutellaria barbata D. Don in adverse-risk acute myeloid leukaemia. Mol Divers 2021; 25:2351-2365. [PMID: 32676746 DOI: 10.1007/s11030-020-10124-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
A poor prognosis, relapse and resistance are burning issues during adverse-risk acute myeloid leukaemia (AML) treatment. As a natural medicine, Scutellaria barbata D. Don (SBD) has shown impressive antitumour activity in various cancers. Thus, SBD may become a potential drug in adverse-risk AML treatment. This study aimed to screen the key targets of SBD in adverse-risk AML using the drug-biomarker interaction model through bioinformatics and network pharmacology methods. First, the adverse-risk AML-related critical biomarkers and targets of SBD active ingredient were obtained from The Cancer Genome Atlas database and several pharmacophore matching databases. Next, the protein-protein interaction network was constructed, and topological analysis and pathway enrichment were used to screen key targets and main pathways of intervention of SBD in adverse-risk AML. Finally, molecular docking was implemented for key target verification. The results suggest that luteolin and quercetin are the main active components of SBD against adverse-risk AML, and affected drug resistance, apoptosis, immune regulation and angiogenesis through the core targets AKT1, MAPK1, IL6, EGFR, SRC, VEGFA and TP53. We hope the proposed drug-biomarker interaction model provides an effective strategy for the research and development of antitumour drugs.
Collapse
Affiliation(s)
- Teng Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong Province, People's Republic of China
| | - Chun-Yi Lyu
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong Province, People's Republic of China
| | - Yue-Hua Jiang
- Central Laboratory of Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan, 250014, Shandong Province, People's Republic of China
| | - Xue-Yan Dong
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong Province, People's Republic of China
| | - Yan Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong Province, People's Republic of China
| | - Zong-Hong Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong Province, People's Republic of China
| | - Jin-Xin Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong Province, People's Republic of China
| | - Rui-Rong Xu
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong Province, People's Republic of China.
| |
Collapse
|
36
|
Pandey P, Khan F, Qari HA, Oves M. Rutin (Bioflavonoid) as Cell Signaling Pathway Modulator: Prospects in Treatment and Chemoprevention. Pharmaceuticals (Basel) 2021; 14:1069. [PMID: 34832851 PMCID: PMC8621917 DOI: 10.3390/ph14111069] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is a complex ailment orchestrated by numerous intrinsic and extrinsic pathways. Recent research has displayed a deep interest in developing plant-based cancer therapeutics for better management of the disease and limited side effects. A wide range of plant-derived compounds have been reported for their anticancer potential in the quest of finding an effective therapeutic approach. Rutin (vitamin P) is a low-molecular weight flavonoid glycoside (polyphenolic compound), abundantly present in various vegetables, fruits (especially berries and citrus fruits), and medicinal herbs. Numerous studies have delineated several pharmacological properties of rutin such as its antiprotozoal, antibacterial, anti-inflammatory, antitumor, antiviral, antiallergic, vasoactive, cytoprotective, antispasmodic, hypolipidemic, antihypertensive, and antiplatelet properties. Specifically, rutin-mediated anticancerous activities have been reported in several cancerous cell lines, but the most common scientific evidence, encompassing several molecular processes and interactions, including apoptosis pathway regulation, aberrant cell signaling pathways, and oncogenic genes, has not been thoroughly studied. In this direction, we attempted to project rutin-mediated oncogenic pathway regulation in various carcinomas. Additionally, we also incorporated advanced research that has uncovered the notable potential of rutin in the modulation of several key cellular functions via interaction with mRNAs, with major emphasis on elucidating direct miRNA targets of rutin as well as the process needed to transform these approaches for developing novel therapeutic interventions for the treatment of several cancers.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida 201306, India;
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida 201306, India;
| | - Huda A. Qari
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mohammad Oves
- Center of Excellence in Environmental Studies, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
37
|
Ashrafizadeh M, Zarrabi A, Mirzaei S, Hashemi F, Samarghandian S, Zabolian A, Hushmandi K, Ang HL, Sethi G, Kumar AP, Ahn KS, Nabavi N, Khan H, Makvandi P, Varma RS. Gallic acid for cancer therapy: Molecular mechanisms and boosting efficacy by nanoscopical delivery. Food Chem Toxicol 2021; 157:112576. [PMID: 34571052 DOI: 10.1016/j.fct.2021.112576] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 07/23/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Cancer is the second leading cause of death worldwide. Majority of recent research efforts in the field aim to address why cancer resistance to therapy develops and how to overcome or prevent it. In line with this, novel anti-cancer compounds are desperately needed for chemoresistant cancer cells. Phytochemicals, in view of their pharmacological activities and capacity to target various molecular pathways, are of great interest in the development of therapeutics against cancer. Plant-derived-natural products have poor bioavailability which restricts their anti-tumor activity. Gallic acid (GA) is a phenolic acid exclusively found in natural sources such as gallnut, sumac, tea leaves, and oak bark. In this review, we report on the most recent research related to anti-tumor activities of GA in various cancers with a focus on its underlying molecular mechanisms and cellular pathwaysthat that lead to apoptosis and migration of cancer cells. GA down-regulates the expression of molecular pathways involved in cancer progression such as PI3K/Akt. The co-administration of GA with chemotherapeutic agents shows improvements in suppressing cancer malignancy. Various nano-vehicles such as organic- and inorganic nano-materials have been developed for targeted delivery of GA at the tumor site. Here, we suggest that nano-vehicles improve GA bioavailability and its ability for tumor suppression.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey; Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Farid Hashemi
- Phd student of pharmacology, Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hui Li Ang
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan.
| | - Pooyan Makvandi
- Centre for Materials Interfaces, Istituto Italiano di Tecnologia, viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy.
| | - Rajender S Varma
- Regional Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| |
Collapse
|
38
|
Liu T, Li Z, Tian F. Quercetin inhibited the proliferation and invasion of hepatoblastoma cells through facilitating SIRT6-medicated FZD4 silence. Hum Exp Toxicol 2021; 40:S96-S107. [PMID: 34219513 DOI: 10.1177/09603271211030558] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hepatoblastoma (HB) is a malignant liver tumor that occurs during childhood. The histone deacetylase SIRT6 functions as a tumor suppressor in diverse cancers. Quercetin, as activators and antioxidants of sirtuins, exhibits remarkable anticancer activity in many tumors. However, whether quercetin ameliorates HB is still unclear. In our study, we found that SIRT6 was downregulated in HB tissues and cell lines. Overexpression of SIRT6 observably suppressed cell proliferation and invasion, promoted cell apoptosis. Mechanistically, SIRT6 suppressed frizzled 4 (FZD4) transcription by deacetylating histone H3K9. Upregulation of SIRT6 reduced the protein levels of FZD4 and H3K9ac. Additionally, quercetin treatment could enhance the expression of SIRT6, repress FZD4 level, cell viability and invasion, and promote apoptosis. Overexpression of FZD4 signally reversed quercetin-treated the promotion effect on cell apoptosis, and the inhibition effects on FZD4 expression, cell viability, invasion and Wnt/β-catenin pathway related proteins. In addition, LiCl, an agonist of Wnt/β-catenin pathway, could recover the inhibition effects of quercetin on Wnt/β-catenin pathway related proteins, cell viability and invasion, and promotion effect on cell apoptosis. In vivo mouse xenograft tumor growth assay revealed that quercetin markedly suppressed tumor growth. In conclusion, these results demonstrated that the molecular mechanism of quercetin suppressing HB cell proliferation and invasion, promoting apoptosis was to promote the deacetylation of SIRT6 on FZD4 and inhibit the activation of Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- T Liu
- The Second Ward of General Surgery Department, Xi'an Children's Hospital, Xi'an, People's Republic of China
| | - Z Li
- The Second Ward of General Surgery Department, Xi'an Children's Hospital, Xi'an, People's Republic of China
| | - F Tian
- The Second Ward of General Surgery Department, Xi'an Children's Hospital, Xi'an, People's Republic of China
| |
Collapse
|
39
|
Lin FJ, Li H, Wu DT, Zhuang QG, Li HB, Geng F, Gan RY. Recent development in zebrafish model for bioactivity and safety evaluation of natural products. Crit Rev Food Sci Nutr 2021; 62:8646-8674. [PMID: 34058920 DOI: 10.1080/10408398.2021.1931023] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zebrafish is a species of freshwater fish, popular in aquariums and laboratories. Several advantageous features have facilitated zebrafish to be extensively utilized as a valuable vertebrate model in the lab. It has been well-recognized that natural products possess multiple health benefits for humans. With the increasing demand for natural products in the development of functional foods, nutraceuticals, and natural cosmetics, the zebrafish has emerged as an unprecedented tool for rapidly and economically screening and identifying safe and effective substances from natural products. This review first summarized the key factors for the management of zebrafish in the laboratory, followed by highlighting the current progress on the establishment and applications of zebrafish models in the bioactivity evaluation of natural products. In addition, the zebrafish models used for assessing the potential toxicity or health risks of natural products were involved as well. Overall, this review indicates that zebrafish are promising animal models for the bioactivity and safety evaluation of natural products, and zebrafish models can accelerate the discovery of novel natural products with potential health functions.
Collapse
Affiliation(s)
- Fang-Jun Lin
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Hang Li
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| | - Ding-Tao Wu
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Qi-Guo Zhuang
- China-New Zealand Belt and Road Joint Laboratory on Kiwifruit, Sichuan Provincial Academy of Natural Resource Sciences, Chengdu, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Ren-You Gan
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| |
Collapse
|
40
|
The Anticancer Effects of Flavonoids through miRNAs Modulations in Triple-Negative Breast Cancer. Nutrients 2021; 13:nu13041212. [PMID: 33916931 PMCID: PMC8067583 DOI: 10.3390/nu13041212] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/31/2022] Open
Abstract
Triple- negative breast cancer (TNBC) incidence rate has regularly risen over the last decades and is expected to increase in the future. Finding novel treatment options with minimum or no toxicity is of great importance in treating or preventing TNBC. Flavonoids are new attractive molecules that might fulfill this promising therapeutic option. Flavonoids have shown many biological activities, including antioxidant, anti-inflammatory, and anticancer effects. In addition to their anticancer effects by arresting the cell cycle, inducing apoptosis, and suppressing cancer cell proliferation, flavonoids can modulate non-coding microRNAs (miRNAs) function. Several preclinical and epidemiological studies indicate the possible therapeutic potential of these compounds. Flavonoids display a unique ability to change miRNAs' levels via different mechanisms, either by suppressing oncogenic miRNAs or activating oncosuppressor miRNAs or affecting transcriptional, epigenetic miRNA processing in TNBC. Flavonoids are not only involved in the regulation of miRNA-mediated cancer initiation, growth, proliferation, differentiation, invasion, metastasis, and epithelial-to-mesenchymal transition (EMT), but also control miRNAs-mediated biological processes that significantly impact TNBC, such as cell cycle, immune system, mitochondrial dysregulation, modulating signaling pathways, inflammation, and angiogenesis. In this review, we highlighted the role of miRNAs in TNBC cancer progression and the effect of flavonoids on miRNA regulation, emphasizing their anticipated role in the prevention and treatment of TNBC.
Collapse
|
41
|
Franza L, Carusi V, Nucera E, Pandolfi F. Luteolin, inflammation and cancer: Special emphasis on gut microbiota. Biofactors 2021; 47:181-189. [PMID: 33507594 DOI: 10.1002/biof.1710] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
Luteolin belongs to the family of flavonoids, which have anti-inflammatory functions, potentially useful in a clinical context, particularly for patients suffering from cancer, neuropsychiatric disorders, inflammatory bowel conditions. This peculiarity has been used for centuries in traditional Chinese medicine, for many different diseases. Its anti-inflammatory effects might be particularly relevant in cancer, with some studies reporting anti-angiogenesis, anti-metastatic, and apoptotic effects on cancer cells by luteolin and other flavonoids. In this article, we analyze the anti-inflammatory role of luteolin, discussing the pathways it may act on. We will then discuss the possible role of microbiota in inflammatory modulation by luteolin. Finally, the possible therapeutic applications of luteolin's anti-inflammatory properties will be analyzed, with a particular focus on cancer.
Collapse
Affiliation(s)
- Laura Franza
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Carusi
- Immunology and Allergy, Internal Medicine Department, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Nucera
- Immunology and Allergy, Internal Medicine Department, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Franco Pandolfi
- Immunology and Allergy, Internal Medicine Department, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
42
|
Almatroodi SA, Alsahli MA, Almatroudi A, Verma AK, Aloliqi A, Allemailem KS, Khan AA, Rahmani AH. Potential Therapeutic Targets of Quercetin, a Plant Flavonol, and Its Role in the Therapy of Various Types of Cancer through the Modulation of Various Cell Signaling Pathways. Molecules 2021; 26:molecules26051315. [PMID: 33804548 PMCID: PMC7957552 DOI: 10.3390/molecules26051315] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
Polyphenolic flavonoids are considered natural, non-toxic chemopreventers, which are most commonly derived from plants, fruits, and vegetables. Most of these polyphenolics exhibit remarkable antioxidant, anti-inflammatory, and anticancer properties. Quercetin (Qu) is a chief representative of these polyphenolic compounds, which exhibits excellent antioxidant and anticancer potential, and has attracted the attention of researchers working in the area of cancer biology. Qu can regulate numerous tumor-related activities, such as oxidative stress, angiogenesis, cell cycle, tumor necrosis factor, proliferation, apoptosis, and metastasis. The anticancer properties of Qu mainly occur through the modulation of vascular endothelial growth factor (VEGF), apoptosis, phosphatidyl inositol-3-kinase (P13K)/Akt (proteinase-kinase B)/mTOR (mammalian target of rapamycin), MAPK (mitogen activated protein kinase)/ERK1/2 (extracellular signal-regulated kinase 1/2), and Wnt/β-catenin signaling pathways. The anticancer potential of Qu is documented in numerous in vivo and in vitro studies, involving several animal models and cell lines. Remarkably, this phytochemical possesses toxic activities against cancerous cells only, with limited toxic effects on normal cells. In this review, we present extensive research investigations aimed to discuss the therapeutic potential of Qu in the management of different types of cancers. The anticancer potential of Qu is specifically discussed by focusing its ability to target specific molecular signaling, such as p53, epidermal growth factor receptor (EGFR), VEGF, signal transducer and activator of transcription (STAT), PI3K/Akt, and nuclear factor kappa B (NF-κB) pathways. The anticancer potential of Qu has gained remarkable interest, but the exact mechanism of its action remains unclear. However, this natural compound has great pharmacological potential; it is now believed to be a complementary—or alternative—medicine for the prevention and treatment of different cancers.
Collapse
Affiliation(s)
- Saleh A. Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia; (S.A.A.); (M.A.A.); (A.A.); (K.S.A.)
| | - Mohammed A. Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia; (S.A.A.); (M.A.A.); (A.A.); (K.S.A.)
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia; (S.A.A.); (M.A.A.); (A.A.); (K.S.A.)
| | - Amit Kumar Verma
- Department of Biotechnology, Jamia Millia Islamia, New Delhi 51542, India;
| | - Abdulaziz Aloliqi
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia;
| | - Khaled S. Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia; (S.A.A.); (M.A.A.); (A.A.); (K.S.A.)
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia;
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia; (S.A.A.); (M.A.A.); (A.A.); (K.S.A.)
- Correspondence:
| |
Collapse
|
43
|
Muoio MG, Talia M, Lappano R, Sims AH, Vella V, Cirillo F, Manzella L, Giuliano M, Maggiolini M, Belfiore A, De Francesco EM. Activation of the S100A7/RAGE Pathway by IGF-1 Contributes to Angiogenesis in Breast Cancer. Cancers (Basel) 2021; 13:cancers13040621. [PMID: 33557316 PMCID: PMC7915817 DOI: 10.3390/cancers13040621] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Breast cancer mortality is increased in patients affected by metabolic disorders associated with dysregulation of the Insulin-like growth factor-1 (IGF-1) axis, like obesity and type-2 diabetes. Despite the oncogenic role of this complex signaling system is widely known, the clinical targeting of IGF-1 and its receptor (IGF-1R) has provided valuable benefit only on small sub-populations of cancer patients, thus suggesting that a further characterization of the biological effects of the IGF-1/IGF-1R pathway could pave the way for a better manipulation of this crucial signaling system at the clinical level. In this study, we have identified the protein S100A7 as novel molecular target of IGF-1 action in the breast tumor microenvironment, toward increased cancer-associated angiogenesis. Targeting the IGF-1/IGF-1R/S100A7 pathway may therefore represent a further useful approach for blocking disease progression in breast cancer patients with dysregulated IGF-1 signaling. Abstract Background: Breast cancer (BC) mortality is increased among obese and diabetic patients. Both obesity and diabetes are associated with dysregulation of both the IGF-1R and the RAGE (Receptor for Advanced Glycation End Products) pathways, which contribute to complications of these disorders. The alarmin S100A7, signaling through the receptor RAGE, prompts angiogenesis, inflammation, and BC progression. Methods: We performed bioinformatic analysis of BC gene expression datasets from published studies. We then used Estrogen Receptor (ER)-positive BC cells, CRISPR-mediated IGF-1R KO BC cells, and isogenic S100A7-transduced BC cells to investigate the role of IGF-1/IGF-1R in the regulation of S100A7 expression and tumor angiogenesis. To this aim, we also used gene silencing and pharmacological inhibitors, and we performed gene expression and promoter studies, western blotting analysis, ChIP and ELISA assays, endothelial cell proliferation and tube formation assay. Results: S100A7 expression correlates with worse prognostic outcomes in human BCs. In BC cells, the IGF-1/IGF-1R signaling engages STAT3 activation and its recruitment to the S100A7 promoter toward S100A7 increase. In human vascular endothelial cells, S100A7 activates RAGE signaling and prompts angiogenic effects. Conclusions: In ER-positive BCs the IGF-1 dependent activation of the S100A7/RAGE signaling in adjacent endothelial cells may serve as a previously unidentified angiocrine effector. Targeting S100A7 may pave the way for a better control of BC, particularly in conditions of unopposed activation of the IGF-1/IGF-1R axis.
Collapse
Affiliation(s)
- Maria Grazia Muoio
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Andrew H. Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh EH4 2XU, UK;
| | - Veronica Vella
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Livia Manzella
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95122 Catania, Italy;
- Department of Clinical and Experimental Medicine, University of Catania, 95122 Catania, Italy
| | - Marika Giuliano
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
- Correspondence: (A.B.); (E.M.D.F.); Tel.: 39-095-7598-700 (A.B.); +39-095-7598-831 (E.M.D.F.)
| | - Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
- Correspondence: (A.B.); (E.M.D.F.); Tel.: 39-095-7598-700 (A.B.); +39-095-7598-831 (E.M.D.F.)
| |
Collapse
|
44
|
Zang X, Cheng M, Zhang X, Chen X. Quercetin nanoformulations: a promising strategy for tumor therapy. Food Funct 2021; 12:6664-6681. [PMID: 34152346 DOI: 10.1039/d1fo00851j] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phytochemicals as dietary constituents are being widely explored for the prevention and treatment of various diseases. Quercetin, a major constituent of various dietary products, has attracted extensive interest due to its anti-proliferative capability, reversal of multidrug resistance, autophagy promotion and tumor microenvironment modulation on different cancer types. Although quercetin has shown potent medical value, its application as an antitumor drug is limited. Problems like poor solubility, bioavailability and stability, short half-life and weak tumor-targeting biodistribution make quercetin an unreliable candidate for cancer therapy. Nanoparticle based platforms have shown a number of advantages in delivering a hydrophobic drug like quercetin to diseased tissues. Quercetin nanoparticles have demonstrated high encapsulation efficiency, stability, sustained release, prolonged circulation time, improved accumulation at tumor sites and therapeutic efficiency. Moreover, a combination of quercetin with other diagnostic or therapeutic agents in one nanocarrier has achieved enhancements in detecting or treating tumors. In this review, we have tried to summarize the pharmacological activities of quercetin with regard to tumor cells and microenvironments in vitro and in vivo. Furthermore, various nanoformulations have been highlighted for quercetin delivery for cancer treatment. These results suggest that quercetin nanoparticles may be a promising antitumor therapeutic agent.
Collapse
Affiliation(s)
- Xinlong Zang
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao, PR China.
| | | | | | | |
Collapse
|
45
|
Systematic Elucidation of the Mechanism of Quercetin against Gastric Cancer via Network Pharmacology Approach. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3860213. [PMID: 32964029 PMCID: PMC7486643 DOI: 10.1155/2020/3860213] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022]
Abstract
This study was aimed at elucidating the potential mechanisms of quercetin in the treatment of gastric cancer (GC). A network pharmacology approach was used to analyze the targets and pathways of quercetin in treating GC. The predicted targets of quercetin against GC were obtained through database mining, and the correlation of these targets with GC was analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Next, the protein-protein interaction (PPI) network was constructed, and overall survival (OS) analysis of hub targets was performed using the Kaplan–Meier Plotter online tool. Finally, the mechanism was further analyzed via molecular docking of quercetin with the hub targets. Thirty-six quercetin-related genes were identified, 15 of which overlapped with GC-related targets. These targets were further mapped to 319 GO biological process terms and 10 remarkable pathways. In the PPI network analysis, six hub targets were identified, including AKT1, EGFR, SRC, IGF1R, PTK2, and KDR. The high expression of these targets was related to poor OS in GC patients. Molecular docking analysis confirmed that quercetin can bind to these hub targets. In conclusion, this study provided a novel approach to reveal the therapeutic mechanisms of quercetin on GC, which will ease the future clinical application of quercetin in the treatment of GC.
Collapse
|
46
|
Imran M, Iqubal MK, Imtiyaz K, Saleem S, Mittal S, Rizvi MMA, Ali J, Baboota S. Topical nanostructured lipid carrier gel of quercetin and resveratrol: Formulation, optimization, in vitro and ex vivo study for the treatment of skin cancer. Int J Pharm 2020; 587:119705. [DOI: 10.1016/j.ijpharm.2020.119705] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/16/2022]
|
47
|
Anti-Cancer Potential of Cannabinoids, Terpenes, and Flavonoids Present in Cannabis. Cancers (Basel) 2020; 12:cancers12071985. [PMID: 32708138 PMCID: PMC7409346 DOI: 10.3390/cancers12071985] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years, and even more since its legalization in several jurisdictions, cannabis and the endocannabinoid system have received an increasing amount of interest related to their potential exploitation in clinical settings. Cannabinoids have been suggested and shown to be effective in the treatment of various conditions. In cancer, the endocannabinoid system is altered in numerous types of tumours and can relate to cancer prognosis and disease outcome. Additionally, cannabinoids display anticancer effects in several models by suppressing the proliferation, migration and/or invasion of cancer cells, as well as tumour angiogenesis. However, the therapeutic use of cannabinoids is currently limited to the treatment of symptoms and pain associated with chemotherapy, while their potential use as cytotoxic drugs in chemotherapy still requires validation in patients. Along with cannabinoids, cannabis contains several other compounds that have also been shown to exert anti-tumorigenic actions. The potential anti-cancer effects of cannabinoids, terpenes and flavonoids, present in cannabis, are explored in this literature review.
Collapse
|
48
|
Flavonoids in Cancer Metastasis. Cancers (Basel) 2020; 12:cancers12061498. [PMID: 32521759 PMCID: PMC7352928 DOI: 10.3390/cancers12061498] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
Metastasis represents a serious complication in the treatment of cancer. Flavonoids are plant secondary metabolites exerting various health beneficiary effects. The effects of flavonoids against cancer are associated not only with early stages of the cancer process, but also with cancer progression and spread into distant sites. Flavonoids showed potent anti-cancer effects against various cancer models in vitro and in vivo, mediated via regulation of key signaling pathways involved in the migration and invasion of cancer cells and metastatic progression, including key regulators of epithelial-mesenchymal transition or regulatory molecules such as MMPs, uPA/uPAR, TGF-β and other contributors of the complex process of metastatic spread. Moreover, flavonoids modulated also the expression of genes associated with the progression of cancer and improved inflammatory status, a part of the complex process involved in the development of metastasis. Flavonoids also documented clear potential to improve the anti-cancer effectiveness of conventional chemotherapeutic agents. Most importantly, flavonoids represent environmentally-friendly and cost-effective substances; moreover, a wide spectrum of different flavonoids demonstrated safety and minimal side effects during long-termed administration. In addition, the bioavailability of flavonoids can be improved by their conjugation with metal ions or structural modifications by radiation. In conclusion, anti-cancer effects of flavonoids, targeting all phases of carcinogenesis including metastatic progression, should be implemented into clinical cancer research in order to strengthen their potential use in the future targeted prevention and therapy of cancer in high-risk individuals or patients with aggressive cancer disease with metastatic potential.
Collapse
|
49
|
Flavonoids in Ageratum conyzoides L. Exert Potent Antitumor Effects on Human Cervical Adenocarcinoma HeLa Cells In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2696350. [PMID: 32461974 PMCID: PMC7218960 DOI: 10.1155/2020/2696350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022]
Abstract
The Ageratum conyzoides L. (A. conyzoides) is commonly used as a traditional medicine, and its antitumor effects have also been studied. However, the functional roles of flavonoids in A. conyzoides in antitumor activities have not been clarified. The present study is aimed at investigating the biological effects of flavonoids in A. conyzoides on human cervical adenocarcinoma. Firstly, we detected that flavonoids in A. conyzoides significantly inhibited the proliferation, invasion, migration, and clonality of human cervical adenocarcinoma HeLa cells in vitro. Furthermore, we found that flavonoids in A. conyzoides induced significant S phase arrest and apoptosis and obviously decreased the intracellular reactive oxygen species (ROS) level in HeLa cells. Finally, we found that flavonoids in A. conyzoides significantly inhibited the HeLa xenograft tumor growth and epithelial-mesenchymal transition (EMT) in vivo. In conclusion, our results demonstrated the obvious antitumor effects of flavonoids in A. conyzoides on HeLa cells, suggesting that flavonoids in A. conyzoides could be provided as a novel therapeutic compound for human cervical adenocarcinoma.
Collapse
|
50
|
Hua X, Zhang H, Jia J, Chen S, Sun Y, Zhu X. Roles of S100 family members in drug resistance in tumors: Status and prospects. Biomed Pharmacother 2020; 127:110156. [PMID: 32335300 DOI: 10.1016/j.biopha.2020.110156] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
Chemotherapy and targeted therapy can significantly improve survival rates in cancer, but multiple drug resistance (MDR) limits the efficacy of these approaches. Understanding the molecular mechanisms underlying MDR is crucial for improving drug efficacy and clinical outcomes of patients with cancer. S100 proteins belong to a family of calcium-binding proteins and have various functions in tumor development. Increasing evidence demonstrates that the dysregulation of various S100 proteins contributes to the development of drug resistance in tumors, providing a basis for the development of predictive and prognostic biomarkers in cancer. Therefore, a combination of biological inhibitors or sensitizers of dysregulated S100 proteins could enhance therapeutic responses. In this review, we provide a detailed overview of the mechanisms by which S100 family members influence resistance of tumors to cancer treatment, with a focus on the development of effective strategies for overcoming MDR.
Collapse
Affiliation(s)
- Xin Hua
- Southeast University Medical College, Nanjing, 210009, China.
| | - Hongming Zhang
- Department of Respiratory Medicine, Yancheng Third People's Hospital, Southeast University Medical College, Yancheng, 224000, China.
| | - Jinfang Jia
- Southeast University Medical College, Nanjing, 210009, China.
| | - Shanshan Chen
- Southeast University Medical College, Nanjing, 210009, China.
| | - Yue Sun
- Southeast University Medical College, Nanjing, 210009, China.
| | - Xiaoli Zhu
- Southeast University Medical College, Nanjing, 210009, China; Department of Respiratory Medicine, Zhongda Hospital of Southeast University Medical College, Nanjing, 210009, China.
| |
Collapse
|