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
|
Figueira MI, Carvalho TMA, Macário-Monteiro J, Cardoso HJ, Correia S, Vaz CV, Duarte AP, Socorro S. The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens. Biomedicines 2024; 12:1636. [PMID: 39200101 PMCID: PMC11351860 DOI: 10.3390/biomedicines12081636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
The role of estrogens in prostate cancer (PCa) is shrouded in mystery, with its actions going from angelic to devilish. The findings by Huggins and Hodges establishing PCa as a hormone-sensitive cancer have provided the basis for using estrogens in therapy. However, despite the clinical efficacy in suppressing tumor growth and the panoply of experimental evidence describing its anticarcinogenic effects, estrogens were abolished from PCa treatment because of the adverse secondary effects. Notwithstanding, research work over the years has continued investigating the effects of estrogens, reporting their pros and cons in prostate carcinogenesis. In contrast with the beneficial therapeutic effects, many reports have implicated estrogens in the disruption of prostate cell fate and tissue homeostasis. On the other hand, epidemiological data demonstrating the lower incidence of PCa in Eastern countries associated with a higher consumption of phytoestrogens support the beneficial role of estrogens in counteracting cancer development. Many studies have investigated the effects of phytoestrogens and the underlying mechanisms of action, which may contribute to developing safe estrogen-based anti-PCa therapies. This review compiles the existing data on the anti- and protumorigenic actions of estrogens and summarizes the anticancer effects of several phytoestrogens, highlighting their promising features in PCa treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sílvia Socorro
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (M.I.F.)
| |
Collapse
|
3
|
Saadh MJ, Mustafa MA, Malathi H, Ahluwalia G, Kaur S, Al-Dulaimi MAAH, Alubiady MHS, Zain Al-Abdeen SH, Shakier HG, Ali MS, Ahmad I, Abosaoda MK. Targeting the pancreatic tumor microenvironment by plant-derived products and their nanoformulations. Med Oncol 2024; 41:201. [PMID: 39001987 DOI: 10.1007/s12032-024-02443-0] [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: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024]
Abstract
Pancreatic cancer remains a significant health issue with limited treatment options. The tumor stroma, a complex environment made up of different cells and proteins, plays a crucial role in tumor growth and chemoresistance. Targeting tumor stroma, consisting of diverse non-tumor cells such as fibroblasts, extracellular matrix (ECM), immune cells, and also pre-vascular cells is encouraging for remodeling solid cancers, such as pancreatic cancer. Remodeling the stroma of pancreas tumors can be suggested as a strategy for reducing resistance to chemo/immunotherapy. Several studies have shown that phytochemicals from plants can affect the tumor environment and have anti-cancer properties. By targeting key pathways involved in stromal activation, phytochemicals may disrupt communication between the tumor and stroma and make tumor cells more sensitive to different treatments. Additionally, phytochemicals have immunomodulatory and anti-angiogenic properties, all of which contribute to their potential in treating pancreatic cancer. This review will provide a detailed look at how phytochemicals impact the tumor stroma and their effects on pancreatic tumor growth, spread, and response to treatment. It will also explore the potential of combining phytochemicals with other treatment options like chemotherapy, immunotherapy, and radiation.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, University of Imam Jaafar AL-Sadiq, Baghdad, Iraq
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Gunveen Ahluwalia
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, 303012, Rajasthan, India
| | - Sumeet Kaur
- Department of Applied Sciences, Chandigarh Engineering Colleges, Chandigarh Group of Colleges, Jhanjeri, 140307, Mohali, Punjab, India
| | | | | | | | | | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
4
|
Auti A, Tathode M, Marino MM, Vitiello A, Ballini A, Miele F, Mazzone V, Ambrosino A, Boccellino M. Nature's weapons: Bioactive compounds as anti-cancer agents. AIMS Public Health 2024; 11:747-772. [PMID: 39416904 PMCID: PMC11474324 DOI: 10.3934/publichealth.2024038] [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: 02/29/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 10/19/2024] Open
Abstract
Cancer represents a major global health burden, prompting continuous research for effective therapeutic strategies. Natural compounds derived from plants have emerged as potential strategies for preventing cancer and treatment because of their inherent pharmacological properties. This comprehensive review aimed to evaluate the therapeutic potential of five key natural compounds: apigenin, quercetin, piperine, curcumin, and resveratrol in cancer prevention and therapy. By examining their molecular mechanisms and preclinical evidence, this review seeks to elucidate their role as potential adjuvants or stand-alone therapies in cancer management. The exploration of natural compounds as cancer therapeutics offers several advantages, including low toxicity, wide availability, and compatibility with conventional chemotherapeutic agents. We highlighted the current understanding of their anticancer mechanisms and clinical applications for advancing personalized cancer care to improve patient outcomes. We discussed the empirical findings from in vitro, in vivo, and clinical studies reporting biological activity and therapeutic efficacy in antioxidant, immunomodulatory, anti-carcinogenic, and chemo-sensitizing modes. Innovative delivery systems and personalized treatment approaches may further enhance their bioavailability and therapeutic utility in a synergistic approach with chemo- and radiotherapeutic disease management. This review underscores the importance of natural compounds in cancer prevention and treatment, promoting a multidisciplinary approach to the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Amogh Auti
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Madhura Tathode
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Maria Michela Marino
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Antonio Vitiello
- Ministry of Health, Directorate-General for Health Prevention, 00144 Rome, Italy
| | - Andrea Ballini
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122, Foggia, Italy
| | - Francesco Miele
- General Surgery Unit, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Valeria Mazzone
- Department of Experimental Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Alessia Ambrosino
- Department of Experimental Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Mariarosaria Boccellino
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| |
Collapse
|
5
|
Zygmunt A, Gubernator J. Metabolism and structure of PDA as the target for new therapies: possibilities and limitations for nanotechnology. Expert Opin Drug Deliv 2024; 21:845-865. [PMID: 38899424 DOI: 10.1080/17425247.2024.2370492] [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: 01/23/2024] [Accepted: 06/17/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION Certainly, pancreatic ductal adenocarcinoma poses one of the greatest challenges in current oncology. The dense extracellular matrix and low vessel density in PDA tumor impede the effective delivery of drugs, primarily due to the short pharmacokinetics of most drugs and potential electrostatic interactions with stroma components. AREA COVERED Owing to the distinctive metabolism of PDA and challenges in accessing nutrients, there is a growing interest in cell metabolism inhibitors as a potential means to inhibit cancer development. However, even if suitable combinations of inhibitors are identified, the question about their administration remains, as the same hindrances that impede effective treatment with conventional drugs will also hinder the delivery of inhibitors. Methods including nanotechnology to increase drugs in PDA penetrations are reviewed and discussed. EXPERT OPINION Pancreatic cancer is one of the most difficult tumors to treat due to the small number of blood vessels, high content of extracellular matrix, and specialized resistance mechanisms of tumor cells. One possible method of treating this tumor is the use of metabolic inhibitors in combinations that show synergy. Despite promising results in in vitro tests, their effect is uncertain due to the tumor's structure. In the case of pancreatic cancer, priming of the tumor tissue is required through the sequential administration of drugs that generate blood vessels, increase blood flow, and enhance vascular permeability and extracellular matrix. The use of drug carriers with a size of 10-30 nm may be crucial in the therapy of this cancer.
Collapse
Affiliation(s)
- Adrianna Zygmunt
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
6
|
Wei Q, Zhang YH. Flavonoids with Anti-Angiogenesis Function in Cancer. Molecules 2024; 29:1570. [PMID: 38611849 PMCID: PMC11013936 DOI: 10.3390/molecules29071570] [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: 03/10/2024] [Revised: 03/23/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
The formation of new blood vessels, known as angiogenesis, significantly impacts the development of multiple types of cancer. Consequently, researchers have focused on targeting this process to prevent and treat numerous disorders. However, most existing anti-angiogenic treatments rely on synthetic compounds and humanized monoclonal antibodies, often expensive or toxic, restricting patient access to these therapies. Hence, the pursuit of discovering new, affordable, less toxic, and efficient anti-angiogenic compounds is imperative. Numerous studies propose that natural plant-derived products exhibit these sought-after characteristics. The objective of this review is to delve into the anti-angiogenic properties exhibited by naturally derived flavonoids from plants, along with their underlying molecular mechanisms of action. Additionally, we summarize the structure, classification, and the relationship between flavonoids with their signaling pathways in plants as anti-angiogenic agents, including main HIF-1α/VEGF/VEGFR2/PI3K/AKT, Wnt/β-catenin, JNK1/STAT3, and MAPK/AP-1 pathways. Nonetheless, further research and innovative approaches are required to enhance their bioavailability for clinical application.
Collapse
Affiliation(s)
- Qiang Wei
- School of Medicine, Anhui Xinhua University, 555 Wangjiang West Road, Hefei 230088, China;
| | | |
Collapse
|
7
|
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
|
8
|
Liao M, Yao D, Wu L, Luo C, Wang Z, Zhang J, Liu B. Targeting the Warburg effect: A revisited perspective from molecular mechanisms to traditional and innovative therapeutic strategies in cancer. Acta Pharm Sin B 2024; 14:953-1008. [PMID: 38487001 PMCID: PMC10935242 DOI: 10.1016/j.apsb.2023.12.003] [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: 07/05/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer reprogramming is an important facilitator of cancer development and survival, with tumor cells exhibiting a preference for aerobic glycolysis beyond oxidative phosphorylation, even under sufficient oxygen supply condition. This metabolic alteration, known as the Warburg effect, serves as a significant indicator of malignant tumor transformation. The Warburg effect primarily impacts cancer occurrence by influencing the aerobic glycolysis pathway in cancer cells. Key enzymes involved in this process include glucose transporters (GLUTs), HKs, PFKs, LDHs, and PKM2. Moreover, the expression of transcriptional regulatory factors and proteins, such as FOXM1, p53, NF-κB, HIF1α, and c-Myc, can also influence cancer progression. Furthermore, lncRNAs, miRNAs, and circular RNAs play a vital role in directly regulating the Warburg effect. Additionally, gene mutations, tumor microenvironment remodeling, and immune system interactions are closely associated with the Warburg effect. Notably, the development of drugs targeting the Warburg effect has exhibited promising potential in tumor treatment. This comprehensive review presents novel directions and approaches for the early diagnosis and treatment of cancer patients by conducting in-depth research and summarizing the bright prospects of targeting the Warburg effect in cancer.
Collapse
Affiliation(s)
- Minru Liao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dahong Yao
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
| | - Lifeng Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chaodan Luo
- Department of Psychology, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhiwen Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jin Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Zhang J, Wang Y, Wang L, You L, Zhang T. Pancreatic ductal adenocarcinoma chemoresistance: From metabolism reprogramming to novel treatment. Chin Med J (Engl) 2024; 137:408-420. [PMID: 37545027 PMCID: PMC10876258 DOI: 10.1097/cm9.0000000000002758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Indexed: 08/08/2023] Open
Abstract
ABSTRACT As pancreatic cancer (PC) is highly malignant, its patients tend to develop metastasis at an early stage and show a poor response to conventional chemotherapies. First-line chemotherapies for PC, according to current guidelines, include fluoropyrimidine- and gemcitabine-based regimens. Accumulating research on drug resistance has shown that biochemical metabolic aberrations in PC, especially those involving glycolysis and glutamine metabolism, are highly associated with chemoresistance. Additionally, lipid metabolism is a major factor in chemoresistance. However, emerging compounds that target these key metabolic pathways have the potential to overcome chemoresistance. This review summarizes how PC develops chemoresistance through aberrations in biochemical metabolism and discusses novel critical targets and pathways within cancer metabolism for new drug research.
Collapse
Affiliation(s)
- Jingcheng Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yutong Wang
- Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lejunzi Wang
- Department of Anaesthesia, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Clinical Immunology Centre, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
10
|
Heydarzadeh S, Moshtaghie AA, Daneshpour M, Hedayati M. The effect of Apigenin on glycometabolism and cell death in an anaplastic thyroid cancer cell line. Toxicol Appl Pharmacol 2023; 475:116626. [PMID: 37437745 DOI: 10.1016/j.taap.2023.116626] [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: 06/11/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
AIMS AND BACKGROUND A more pronounced characteristic of cancer cells is the energy dependence on glucose, which mitigated by glucose transporters. The comprehension of the regulatory mechanisms behind the Warburg effect holds promise for developing therapeutic interventions for cancers. Studies are lacking which targeted the GLUTs for treatment of malignancy of thyroid tumors. In our current investigation, we have undertaken this study to determine the potential of Apigenin, plant derived flavonoid in modulating tumor apoptosis by targeting GLUTs expression in SW1736 cell line of anaplastic thyroid carcinoma. MATERIAL METHODS Flow cytometry with propidium iodide staining was used to determine cell apoptosis. For glucose uptake detection, the "GOD-PAP" enzymatic colorimetric test was used to measure the direct glucose levels inside the cells. To determine the expression of GLUT1 and GLUT3 mRNA in the SW1736 cell line qRT-PCR was employed. Protein levels of GLUT1 and GLUT3 in the SW1736 cell line were detected with western blotting. Also, the scratch wound healing assay was conducted for cell migration. RESULTS According to qRT-PCR analysis, the levels of GLUT1 and GLUT3 mRNA were lower in the group that received Apigenin relative to the control group. The Apigenin treatment of SW1736 cells decreased protein expression of the GLUT1 and GLUT3 levels in conformity to qRT-PCR. The scratch assays revealed that Apigenin treatment of cancer cell lines inhibited cell migration as compared to control. CONCLUSION These findings demonstrate the possibility of targeting the glucose facilitators' pathway for making thyroid cancer cells more susceptible to programmed cell death.
Collapse
Affiliation(s)
- Shabnam Heydarzadeh
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Ali Asghar Moshtaghie
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Isfahan, Iran.
| | - Maryam Daneshpour
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Abstract
Hypoxia (oxygen deprivation) occurs in most solid malignancies, albeit with considerable heterogeneity. Hypoxia is associated with an aggressive cancer phenotype by promotion of genomic instability, evasion of anti-cancer therapies including radiotherapy and enhancement of metastatic risk. Therefore, hypoxia results in poor cancer outcomes. Targeting hypoxia to improve cancer outcomes is an attractive therapeutic strategy. Hypoxia-targeted dose painting escalates radiotherapy dose to hypoxic sub-volumes, as quantified and spatially mapped using hypoxia imaging. This therapeutic approach could overcome hypoxia-induced radioresistance and improve patient outcomes without the need for hypoxia-targeted drugs. This article will review the premise and underpinning evidence for personalized hypoxia-targeted dose painting. It will present data on relevant hypoxia imaging biomarkers, highlight the challenges and potential benefit of this approach and provide recommendations for future research priorities in this field. Personalized hypoxia-based radiotherapy de-escalation strategies will also be addressed.
Collapse
Affiliation(s)
- Ahmed Salem
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, Hashemite University, Zarqa, Jordan; Division of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
12
|
Moslehi M, Rezaei S, Talebzadeh P, Ansari MJ, Jawad MA, Jalil AT, Rastegar-Pouyani N, Jafarzadeh E, Taeb S, Najafi M. Apigenin in cancer therapy: Prevention of genomic instability and anticancer mechanisms. Clin Exp Pharmacol Physiol 2023; 50:3-18. [PMID: 36111951 DOI: 10.1111/1440-1681.13725] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/18/2022] [Accepted: 09/14/2022] [Indexed: 12/13/2022]
Abstract
The incidence of cancer has been growing worldwide. Better survival rates following the administration of novel drugs and new combination therapies may concomitantly cause concern regarding the long-term adverse effects of cancer therapy, for example, second primary malignancies. Moreover, overcoming tumour resistance to anticancer agents has been long considered as a critical challenge in cancer research. Some low toxic adjuvants such as herb-derived molecules may be of interest for chemoprevention and overcoming the resistance of malignancies to cancer therapy. Apigenin is a plant-derived molecule with attractive properties for chemoprevention, for instance, promising anti-tumour effects, which may make it a desirable adjuvant to reduce genomic instability and the risks of second malignancies among normal tissues. Moreover, it may improve the efficiency of anticancer modalities. This paper aims to review various effects of apigenin in both normal tissues and malignancies. In addition, we explain how apigenin may have the ability to protect usual cells against the genotoxic repercussions following radiotherapy and chemotherapy. Furthermore, the inhibitory effects of apigenin on tumours will be discussed.
Collapse
Affiliation(s)
- Masoud Moslehi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sepideh Rezaei
- Department of Chemistry, University of Houston, Houston, Texas, USA
| | - Pourya Talebzadeh
- Student Research Committee, Tehran Medical Faculty, Islamic Azad University, Tehran, Iran
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
| | | | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, Iraq
| | - Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Emad Jafarzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran.,Medical Biotechnology Research Center, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
13
|
Cui Y, Li C, Sang F, Cao W, Qin Z, Zhang P. Natural products targeting glycolytic signaling pathways-an updated review on anti-cancer therapy. Front Pharmacol 2022; 13:1035882. [PMID: 36339566 PMCID: PMC9631946 DOI: 10.3389/fphar.2022.1035882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
Glycolysis is a complex metabolic process that occurs to convert glucose into pyruvate to produce energy for living cells. Normal cells oxidized pyruvate into adenosine triphosphate and carbon dioxide in the presence of oxygen in mitochondria while cancer cells preferentially metabolize pyruvate to lactate even in the presence of oxygen in order to maintain a slightly acidic micro-environment of PH 6.5 and 6.9, which is beneficial for cancer cell growth and metastasis. Therefore targeting glycolytic signaling pathways provided new strategy for anti-cancer therapy. Natural products are important sources for the treatment of diseases with a variety of pharmacologic activities. Accumulated studies suggested that natural products exhibited remarkable anti-cancer properties both in vitro and in vivo. Plenty of studies suggested natural products like flavonoids, terpenoids and quinones played anti-cancer properties via inhibiting glucose metabolism targets in glycolytic pathways. This study provided an updated overview of natural products controlling glycolytic pathways, which also provide insight into druggable mediators discovery targeting cancer glucose metabolism.
Collapse
Affiliation(s)
- Yuting Cui
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Chuang Li
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Feng Sang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Weiling Cao
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Zhuo Qin
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Peng Zhang
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| |
Collapse
|
14
|
Targeting Glucose Metabolism Enzymes in Cancer Treatment: Current and Emerging Strategies. Cancers (Basel) 2022; 14:cancers14194568. [PMID: 36230492 PMCID: PMC9559313 DOI: 10.3390/cancers14194568] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Reprogramming of glucose metabolism is a hallmark of cancer and can be targeted by therapeutic agents. Some metabolism regulators, such as ivosidenib and enasidenib, have been approved for cancer treatment. Currently, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Furthermore, some natural products have shown efficacy in killing tumor cells by regulating glucose metabolism, offering novel therapeutic opportunities in cancer. However, most of them have failed to be translated into clinical applications due to low selectivity, high toxicity, and side effects. Recent studies suggest that combining glucose metabolism modulators with chemotherapeutic drugs, immunotherapeutic drugs, and other conventional anticancer drugs may be a future direction for cancer treatment. Abstract Reprogramming of glucose metabolism provides sufficient energy and raw materials for the proliferation, metastasis, and immune escape of cancer cells, which is enabled by glucose metabolism-related enzymes that are abundantly expressed in a broad range of cancers. Therefore, targeting glucose metabolism enzymes has emerged as a promising strategy for anticancer drug development. Although several glucose metabolism modulators have been approved for cancer treatment in recent years, some limitations exist, such as a short half-life, poor solubility, and numerous adverse effects. With the rapid development of medicinal chemicals, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Additionally, several studies have found that some natural products can suppress cancer progression by regulating glucose metabolism enzymes. In this review, we summarize the mechanisms underlying the reprogramming of glucose metabolism and present enzymes that could serve as therapeutic targets. In addition, we systematically review the existing drugs targeting glucose metabolism enzymes, including small-molecule modulators and natural products. Finally, the opportunities and challenges for glucose metabolism enzyme-targeted anticancer drugs are also discussed. In conclusion, combining glucose metabolism modulators with conventional anticancer drugs may be a promising cancer treatment strategy.
Collapse
|
15
|
Phytochemicals as Regulators of Tumor Glycolysis and Hypoxia Signaling Pathways: Evidence from In Vitro Studies. Pharmaceuticals (Basel) 2022; 15:ph15070808. [PMID: 35890106 PMCID: PMC9315613 DOI: 10.3390/ph15070808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
The full understanding of the complex nature of cancer still faces many challenges, as cancers arise not as a result of a single target disruption but rather involving successive genetic and epigenetic alterations leading to multiple altered metabolic pathways. In this light, the need for a multitargeted, safe and effective therapy becomes essential. Substantial experimental evidence upholds the potential of plant-derived compounds to interfere in several important pathways, such as tumor glycolysis and the upstream regulating mechanisms of hypoxia. Herein, we present a comprehensive overview of the natural compounds which demonstrated, in vitro studies, an effective anticancer activity by affecting key regulators of the glycolytic pathway such as glucose transporters, hexokinases, phosphofructokinase, pyruvate kinase or lactate dehydrogenase. Moreover, we assessed how phytochemicals could interfere in HIF-1 synthesis, stabilization, accumulation, and transactivation, emphasizing PI3K/Akt/mTOR and MAPK/ERK pathways as important signaling cascades in HIF-1 activation. Special consideration was given to cell culture-based metabolomics as one of the most sensitive, accurate, and comprising approaches for understanding the response of cancer cell metabolome to phytochemicals.
Collapse
|
16
|
Hypoxia as a Modulator of Inflammation and Immune Response in Cancer. Cancers (Basel) 2022; 14:cancers14092291. [PMID: 35565420 PMCID: PMC9099524 DOI: 10.3390/cancers14092291] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
A clear association between hypoxia and cancer has heretofore been established; however, it has not been completely developed. In this sense, the understanding of the tumoral microenvironment is critical to dissect the complexity of cancer, including the reduction in oxygen distribution inside the tumoral mass, defined as tumoral hypoxia. Moreover, hypoxia not only influences the tumoral cells but also the surrounding cells, including those related to the inflammatory processes. In this review, we analyze the participation of HIF, NF-κB, and STAT signaling pathways as the main components that interconnect hypoxia and immune response and how they modulate tumoral growth. In addition, we closely examine the participation of the immune cells and how they are affected by hypoxia, the effects of the progression of cancer, and some innovative applications that take advantage of this knowledge, to suggest potential therapies. Therefore, we contribute to the understanding of the complexity of cancer to propose innovative therapeutic strategies in the future.
Collapse
|
17
|
Bao Y, Zhong J, Shen L, Dai L, Zhou S, Fan J, Yao H, Lu Z. Effect of Glut-1 and HIF-1α double knockout by CRISPR/CAS9 on radiosensitivity in laryngeal carcinoma via the PI3K/Akt/mTOR pathway. J Cell Mol Med 2022; 26:2881-2894. [PMID: 35415942 PMCID: PMC9907005 DOI: 10.1111/jcmm.17303] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 03/13/2022] [Accepted: 03/16/2022] [Indexed: 12/16/2022] Open
Abstract
Hypoxic resistance is the main obstacle to radiotherapy for laryngeal carcinoma. Our previous study indicated that hypoxia-inducible factor 1α (HIF-1α) and glucose transporter 1 (Glut-1) double knockout reduced tumour biological behaviour in laryngeal carcinoma cells. However, their radioresistance mechanism remains unclear. In this study, cell viability was determined by CCK8 assay. Glucose uptake capability was evaluated by measurement of 18 F-fluorodeoxyglucose radioactivity. A tumour xenograft model was established by subcutaneous injection of Tu212 cells. Tumour histopathology was determined by haematoxylin and eosin staining, immunohistochemical staining, and TUNEL assays. Signalling transduction was evaluated by Western blotting. We found that hypoxia induced radioresistance in Tu212 cells accompanied by increased glucose uptake capability and activation of the PI3K/Akt/mTOR pathway. Inhibition of PI3K/Akt/mTOR activity abolished hypoxia-induced radioresistance and glucose absorption. Mechanistic analysis revealed that hypoxia promoted higher expressions of HIF-1α and Glut-1. Moreover, the PI3K/Akt/mTOR pathway was a positive mediator of HIF-1α and/or Glut-1 in the presence of irradiation. HIF-1α and/or Glut-1 knockout significantly reduced cell viability, glucose uptake and PI3K/Akt/mTOR activity, all of which were induced by hypoxia in the presence of irradiation. In vivo analysis showed that knockout of HIF-1α and/or Glut-1 also inhibited tumour growth by promoting cell apoptosis, more robustly compared with the PI3K inhibitor wortmannin, particularly in tumours with knockout of both HIF-1α and Glut-1. HIF-1α and/or Glut-1 knockout also abrogated PI3K/Akt/mTOR signalling transduction in tumour tissues, in a manner similar to wortmannin. HIF-1α and/or Glut-1 knockout facilitated radiosensitivity in laryngeal carcinoma Tu212 cells by regulation of the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Yang‐Yang Bao
- Department of OtolaryngologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou CityChina
| | - Jiang‐Tao Zhong
- Department of OtolaryngologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou CityChina
| | - Li‐Fang Shen
- Department of OtolaryngologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou CityChina
| | - Li‐Bo Dai
- Department of OtolaryngologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou CityChina
| | - Shui‐Hong Zhou
- Department of OtolaryngologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou CityChina
| | - Jun Fan
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhou CityChina
| | - Hong‐Tian Yao
- Department of PathologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou CityChina
| | - Zhong‐Jie Lu
- Department of RadiotherapyThe First Affiliated HospitalZhejiang University School of MedicineHangzhou CityChina
| |
Collapse
|
18
|
Lee HH, Cho H. Apigenin Increases Natural Killer Cytotoxicity to Human Hepatocellular Carcinoma Expressing HIF-1α through High Interaction of CD95/CD95L. J Microbiol Biotechnol 2022; 32:397-404. [PMID: 35283421 PMCID: PMC9628789 DOI: 10.4014/jmb.2201.01010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
Abstract
Natural killer (NK) cell activity is more attenuated in hepatocellular carcinoma (HCC) patients than normal. Hypoxic-inducible factor (HIF)-1α is highly expressed in tumors to maintain their metabolism in a hypoxic environment. The expression of HIF-1α in cancers can lead to cell growth, proliferation, invasion/metastasis and immune escape. Although apigenin, a flavonoid, is known to have various biological activities, it has not been demonstrated in NK cell immune activity in HCC cells. In this study, NK-92 cells were directly cocultured with HCC SK-Hep1 cells for 24 h to evaluate NK cell activity in HCC cells or HCC cells expressing HIF-1α by apigenin. NK cell cytotoxicity to HCC cells expressing HIF-1α was significantly increased, and NK cell-activating receptors, NKG2D, NKp30 and NKp44 were highly expressed. The activating effect of apigenin on NK cells substantially induced apoptosis in HCC cells expressing HIF-1α through high expression of CD95L on the surface of NK-92 cells. Moreover, apigenin excellently inhibited the level of TGF-β1 in a coculture of NK cells and HCC cells. In conclusion, apigenin seems to be a good compound that increases NK cell cytotoxicity to HCC cells by controlling HIF-1α expression.
Collapse
Affiliation(s)
- Hwan Hee Lee
- Department of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea,Duksung Innovative Drug Center, Duksung Women’s University, Seoul 01369, Republic of Korea,Corresponding authors H.H. Lee Phone: +82-2-901-8734 Fax: +82-2-901-8386 E-mail:
| | - Hyosun Cho
- Department of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea,Duksung Innovative Drug Center, Duksung Women’s University, Seoul 01369, Republic of Korea,
H. Cho Phone: +82-2-901-8678 Fax: +82-2-901-8386 E-mail:
| |
Collapse
|
19
|
Chang X, Zhou F, Bu L, Wang N, Deng J, Wang S. Semaphorin 3A attenuates the hypoxia suppression of osteogenesis in periodontal ligament stem cells. J Periodontal Res 2022; 57:425-433. [PMID: 35037251 DOI: 10.1111/jre.12973] [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: 07/29/2021] [Revised: 12/25/2021] [Accepted: 01/06/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE AND BACKGROUND The occurrence and development of periodontitis are closely related to hypoxia of the periodontal microenvironment. Periodontal ligament stem cells (PDLSCs) are considered to have potential to regenerate periodontal tissues. Semaphorin 3A (Sema3A) plays an essential role in promoting osteogenesis. However, the effect of Sema3A on osteogenesis of PDLSCs under hypoxia remains unclear. The aim of this study was to investigate the effect of Sema3A on osteogenesis of PDLSCs under hypoxia. METHODS Isolated PDLSCs were identified using flow cytometry. Adipogenic differentiation potential was identified by oil red O staining. Osteogenesis was measured using Alizarin Red S staining and ALP staining. Intracellular hypoxia was induced using cobalt chloride (CoCl2 ). The expression level of hypoxia-inducible factor-1α (HIF-1α) was detected via ELISA. Expression of osteogenic markers and Sema3A was analyzed using western blot and real-time PCR. RESULTS The proliferation and osteogenesis of PDLSCs were markedly inhibited with increased concentrations of CoCl2 . Under the treatment with a low concentration of CoCl2 , expression of related osteogenic markers and Sema3A decreased in a time-dependent manner. ARS and ALP staining results also showed that osteogenic calcification decreased under hypoxia. Apigenin, an inhibitor of HIF-1α, effectively up-regulated expression of Sema3A and osteogenic markers with CoCl2 treatment. Moreover, exogenous Sema3A significantly increased the expression of osteogenesis-related markers and mineralization of PDLSCs according to ALP and ARS staining with CoCl2 treatment. CONCLUSIONS Hypoxia markedly inhibited osteogenesis of PDLSCs. Sema3A explicitly attenuated the hypoxia suppression of osteogenesis in PDLSCs.
Collapse
Affiliation(s)
- Xiaochi Chang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
| | - Fengyi Zhou
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Lingxue Bu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Nan Wang
- School of Stomatology of Qingdao University, Qingdao, China.,Department of Stomatology, Hospital of the PLA Navy, Qingdao, China
| | - Jing Deng
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
| | - Shuai Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
| |
Collapse
|
20
|
Periyasamy L, Muruganantham B, Park WY, Muthusami S. Phyto-targeting the CEMIP Expression as a Strategy to Prevent Pancreatic Cancer Metastasis. Curr Pharm Des 2022; 28:922-946. [PMID: 35236267 DOI: 10.2174/1381612828666220302153201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Metastasis of primary pancreatic cancer (PC) to adjacent or distant organs is responsible for the poor survival rate of affected individuals. Chemotherapy, radiotherapy, and immunotherapy are currently being prescribed to treat PC in addition to surgical resection. Surgical resection is the preferred treatment for PC that leads to 20% of 5-year survival, but only less than 20% of patients are eligible for surgical resection because of the poor prognosis. To improve the prognosis and clinical outcome, early diagnostic markers need to be identified, and targeting them would be of immense benefit to increase the efficiency of the treatment. Cell migration-inducing hyaluronan-binding protein (CEMIP) is identified as an important risk factor for the metastasis of various cancers, including PC. Emerging studies have pointed out the crucial role of CEMIP in the regulation of various signaling mechanisms, leading to enhanced migration and metastasis of PC. METHODS The published findings on PC metastasis, phytoconstituents, and CEMIP were retrieved from Pubmed, ScienceDirect, and Cochrane Library. Computational tools, such as gene expression profiling interactive analysis (GEPIA) and Kaplan-Meier (KM) plotter, were used to study the relationship between CEMIP expression and survival of PC individuals. RESULTS Gene expression analysis using the GEPIA database identified a stupendous increase in the CEMIP transcript in PC compared to adjacent normal tissues. KM plotter analysis revealed the impact of CEMIP on the overall survival (OS) and disease-free survival (DFS) among PC patients. Subsequently, several risk factors associated with PC development were screened, and their ability to regulate CEMIP gene expression was analyzed using computational tools. CONCLUSION The current review is focused on gathering information regarding the regulatory role of phytocomponents in PC migration and exploring their possible impact on the CEMIP expression.
Collapse
Affiliation(s)
- Loganayaki Periyasamy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641 021, India
| | - Bharathi Muruganantham
- Karpagam Cancer Research Centre, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641 021, India
| | - Woo-Yoon Park
- Department of Radiation Oncology, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641 021, India
- Karpagam Cancer Research Centre, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641 021, India
| |
Collapse
|
21
|
Kim TW, Lee HG. Apigenin Induces Autophagy and Cell Death by Targeting EZH2 under Hypoxia Conditions in Gastric Cancer Cells. Int J Mol Sci 2021; 22:ijms222413455. [PMID: 34948250 PMCID: PMC8706813 DOI: 10.3390/ijms222413455] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 01/17/2023] Open
Abstract
Hypoxia is a major obstacle to gastric cancer (GC) therapy and leads to chemoresistance as GC cells are frequently exposed to the hypoxia environment. Apigenin, a flavonoid found in traditional medicine, fruits, and vegetables and an HDAC inhibitor, is a powerful anti-cancer agent against various cancer cell lines. However, detailed mechanisms involved in the treatment of GC using APG are not fully understood. In this study, we investigated the biological activity of and molecular mechanisms involved in APG-mediated treatment of GC under hypoxia. APG promoted autophagic cell death by increasing ATG5, LC3-II, and phosphorylation of AMPK and ULK1 and down-regulating p-mTOR and p62 in GC. Furthermore, our results show that APG induces autophagic cell death via the activation of the PERK signaling, indicating an endoplasmic reticulum (ER) stress response. The inhibition of ER stress suppressed APG-induced autophagy and conferred prolonged cell survival, indicating autophagic cell death. We further show that APG induces ER stress- and autophagy-related cell death through the inhibition of HIF-1α and Ezh2 under normoxia and hypoxia. Taken together, our findings indicate that APG activates autophagic cell death by inhibiting HIF-1α and Ezh2 under hypoxia conditions in GC cells.
Collapse
Affiliation(s)
- Tae Woo Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: (T.W.K.); (H.G.L.); Tel.: +82-2-961-0329 (T.W.K.); +82-42-860-4182 (H.G.L.); Fax: +82-2-961-1165 (T.W.K.); +82-42-860-4593 (H.G.L.)
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: (T.W.K.); (H.G.L.); Tel.: +82-2-961-0329 (T.W.K.); +82-42-860-4182 (H.G.L.); Fax: +82-2-961-1165 (T.W.K.); +82-42-860-4593 (H.G.L.)
| |
Collapse
|
22
|
Carvalho TMA, Di Molfetta D, Greco MR, Koltai T, Alfarouk KO, Reshkin SJ, Cardone RA. Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches. Cancers (Basel) 2021; 13:6135. [PMID: 34885243 PMCID: PMC8657427 DOI: 10.3390/cancers13236135] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, the median overall survival of PDAC patients rarely exceeds 1 year and has an overall 5-year survival rate of about 9%. These numbers are anticipated to worsen in the future due to the lack of understanding of the factors involved in its strong chemoresistance. Chemotherapy remains the only treatment option for most PDAC patients; however, the available therapeutic strategies are insufficient. The factors involved in chemoresistance include the development of a desmoplastic stroma which reprograms cellular metabolism, and both contribute to an impaired response to therapy. PDAC stroma is composed of immune cells, endothelial cells, and cancer-associated fibroblasts embedded in a prominent, dense extracellular matrix associated with areas of hypoxia and acidic extracellular pH. While multiple gene mutations are involved in PDAC initiation, this desmoplastic stroma plays an important role in driving progression, metastasis, and chemoresistance. Elucidating the mechanisms underlying PDAC resistance are a prerequisite for designing novel approaches to increase patient survival. In this review, we provide an overview of the stromal features and how they contribute to the chemoresistance in PDAC treatment. By highlighting new paradigms in the role of the stromal compartment in PDAC therapy, we hope to stimulate new concepts aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Tiago M. A. Carvalho
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Maria Raffaella Greco
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | | | - Khalid O. Alfarouk
- Al-Ghad International College for Applied Medical Sciences, Al-Madinah Al-Munwarah 42316, Saudi Arabia;
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Rosa A. Cardone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| |
Collapse
|
23
|
Sarigul Sezenoz A, Akkoyun I, Helvacioglu F, Haberal N, Dagdeviren A, Bacanli D, Yilmaz G, Oto S. Antiproliferative and Mitochondrial Protective Effects of Apigenin in an Oxygen-Induced Retinopathy In Vivo Mouse Model. J Ocul Pharmacol Ther 2021; 37:580-590. [PMID: 34665015 DOI: 10.1089/jop.2021.0046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Purpose: To investigate the effects of a common dietary flavonoid apigenin on retinal endothelial cell proliferation, retinal morphological structure, and apoptotic cell death in an oxygen-induced retinopathy (OIR) mouse model to evaluate the possibility of the use of apigenin in the treatment of ocular neovascular diseases (ONDs). Methods: Ninety-six newborn C57BL/6J mice were included. Eight groups were randomized, each including 12 mice. Two negative control groups were kept in room air: the first without any injection and the second received intravitreal (IV) dimethyl sulfoxide (DMSO), which is the solvent we used. The OIR groups were exposed to 75% ± 2% oxygen from postnatal days (PD) 7 to 12. On PD 12, the mice were randomly assigned to 6 groups: 2 OIR control groups (1 received no injection, 1 received IV-DMSO), 2 IV-apigenin groups (10 and 20 μg/mL), and 2 intraperitoneal (IP)-apigenin groups (10 and 20 mg/kg). We quantified retinal endothelial cell proliferation by counting neovascular tufts in cross-sections and examined histological and ultrastructural changes through light and electron microscopy. We evaluated apoptosis by terminal deoxynucleotidyl transferase-mediated nick end-labeling (TUNEL). Results: We detected a significant increase in endothelial cell proliferation in the OIR groups. Groups receiving apigenin, both IP and IV, had significant decreases in endothelial cells, atypical mitochondrion count, and apoptotic cells compared with the groups receiving no injections. None of the apigenin-injected groups revealed cystic degeneration or cell loss. Conclusions: Apigenin suppresses neovascularization, has antiapoptotic and antioxidative effects in an OIR mouse model, and can be considered a promising agent for treating OND. Clinical trial (Project number: DA15/19).
Collapse
Affiliation(s)
| | - Imren Akkoyun
- Department of Ophthalmology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Fatma Helvacioglu
- Department of Histology and Embryology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Nihan Haberal
- Department of Pathology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Attila Dagdeviren
- Department of Histology and Embryology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Didem Bacanli
- Baskent University Laboratory Animal Breeding and Research Center, Ankara, Turkey
| | - Gursel Yilmaz
- Department of Ophthalmology, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Sibel Oto
- Department of Ophthalmology, Baskent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
24
|
Rashid M, Zadeh LR, Baradaran B, Molavi O, Ghesmati Z, Sabzichi M, Ramezani F. Up-down regulation of HIF-1α in cancer progression. Gene 2021; 798:145796. [PMID: 34175393 DOI: 10.1016/j.gene.2021.145796] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/25/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022]
Abstract
Hypoxia induicible factor-1 alpha (HIF-1α) is a key transcription factor in cancer progression and target therapy in cancer. HIF-1α acts differently depending on presence or absence of Oxygen. In an oxygen-immersed environment, HIF-1α completely deactivated and destroyed by the ubiquitin proteasome pathway (UPP). In contrast, in the oxygen-free environment, it escapes destruction and enters to the nucleus of cells then upregulates many genes involved in cancer progression. Overexpressed HIF-1α and downstream genes support cancer progression through various mechanisms including angiogenesis, proliferation and survival of cells, metabolism reprogramming, invasion and metastasis, cancer stem cell maintenance, induction of genetic instability, and treatment resistance. HIF-1α can be provoked by signaling pathways unrelated to hypoxia during cancer progression. Therefore, cancer development and progression can be modulated by targeting HIF-1α and its downstream signaling molecules. In this regard, HIF-1α inhibitors which are categorized into the agents that regulate HIF-1α in gene, mRNA and protein levels used as an efficient way in cancer treatment. Also, HIF-1α expression can be negatively affected by the agents suppressing the activation of mTOR, PI3k/Akt and MAPK pathways.
Collapse
Affiliation(s)
- Mohsen Rashid
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Rostami Zadeh
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Ghesmati
- Department of Medical Biotechnology, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Sabzichi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Fatemeh Ramezani
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
25
|
Shirai Y, Chow CCT, Kambe G, Suwa T, Kobayashi M, Takahashi I, Harada H, Nam JM. An Overview of the Recent Development of Anticancer Agents Targeting the HIF-1 Transcription Factor. Cancers (Basel) 2021; 13:cancers13112813. [PMID: 34200019 PMCID: PMC8200185 DOI: 10.3390/cancers13112813] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia, a characteristic feature of solid tumors, is associated with the malignant phenotype and therapy resistance of cancers. Hypoxia-inducible factor 1 (HIF-1), which is responsible for the metazoan adaptive response to hypoxia, has been recognized as a rational target for cancer therapy due to its critical functions in hypoxic regions. In order to efficiently inhibit its activity, extensive efforts have been made to elucidate the molecular mechanism underlying the activation of HIF-1. Here, we provide an overview of relevant research, particularly on a series of HIF-1 activators identified so far and the development of anticancer drugs targeting them.
Collapse
Affiliation(s)
- Yukari Shirai
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Christalle C. T. Chow
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Gouki Kambe
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Tatsuya Suwa
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Itsuki Takahashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
- Correspondence: (H.H.); (J.-M.N.); Tel.: +81-75-753-7560 (H.H.); +81-75-753-7567 (J.-M.N.)
| | - Jin-Min Nam
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan; (Y.S.); (C.C.T.C.); (G.K.); (T.S.); (M.K.); (I.T.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-Cho, Sakyo-Ku, Kyoto 606-8501, Japan
- Correspondence: (H.H.); (J.-M.N.); Tel.: +81-75-753-7560 (H.H.); +81-75-753-7567 (J.-M.N.)
| |
Collapse
|
26
|
Ponte LGS, Pavan ICB, Mancini MCS, da Silva LGS, Morelli AP, Severino MB, Bezerra RMN, Simabuco FM. The Hallmarks of Flavonoids in Cancer. Molecules 2021; 26:2029. [PMID: 33918290 PMCID: PMC8038160 DOI: 10.3390/molecules26072029] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Flavonoids represent an important group of bioactive compounds derived from plant-based foods and beverages with known biological activity in cells. From the modulation of inflammation to the inhibition of cell proliferation, flavonoids have been described as important therapeutic adjuvants against several diseases, including diabetes, arteriosclerosis, neurological disorders, and cancer. Cancer is a complex and multifactor disease that has been studied for years however, its prevention is still one of the best known and efficient factors impacting the epidemiology of the disease. In the molecular and cellular context, some of the mechanisms underlying the oncogenesis and the progression of the disease are understood, known as the hallmarks of cancer. In this text, we review important molecular signaling pathways, including inflammation, immunity, redox metabolism, cell growth, autophagy, apoptosis, and cell cycle, and analyze the known mechanisms of action of flavonoids in cancer. The current literature provides enough evidence supporting that flavonoids may be important adjuvants in cancer therapy, highlighting the importance of healthy and balanced diets to prevent the onset and progression of the disease.
Collapse
Affiliation(s)
- Luis Gustavo Saboia Ponte
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
- Laboratory of Signal Mechanisms (LMS), School of Pharmaceutical Sciences (FCF), University of Campinas (UNICAMP), Campinas, São Paulo 13083-871, Brazil
| | - Mariana Camargo Silva Mancini
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Luiz Guilherme Salvino da Silva
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Ana Paula Morelli
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Matheus Brandemarte Severino
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Rosangela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil; (L.G.S.P.); (I.C.B.P.); (M.C.S.M.); (L.G.S.d.S.); (A.P.M.); (M.B.S.); (R.M.N.B.)
| |
Collapse
|
27
|
Samec M, Liskova A, Koklesova L, Mersakova S, Strnadel J, Kajo K, Pec M, Zhai K, Smejkal K, Mirzaei S, Hushmandi K, Ashrafizadeh M, Saso L, Brockmueller A, Shakibaei M, Büsselberg D, Kubatka P. Flavonoids Targeting HIF-1: Implications on Cancer Metabolism. Cancers (Basel) 2021; 13:E130. [PMID: 33401572 PMCID: PMC7794792 DOI: 10.3390/cancers13010130] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor hypoxia is described as an oxygen deprivation in malignant tissue. The hypoxic condition is a consequence of an imbalance between rapidly proliferating cells and a vascularization that leads to lower oxygen levels in tumors. Hypoxia-inducible factor 1 (HIF-1) is an essential transcription factor contributing to the regulation of hypoxia-associated genes. Some of these genes modulate molecular cascades associated with the Warburg effect and its accompanying pathways and, therefore, represent promising targets for cancer treatment. Current progress in the development of therapeutic approaches brings several promising inhibitors of HIF-1. Flavonoids, widely occurring in various plants, exert a broad spectrum of beneficial effects on human health, and are potentially powerful therapeutic tools against cancer. Recent evidences identified numerous natural flavonoids and their derivatives as inhibitors of HIF-1, associated with the regulation of critical glycolytic components in cancer cells, including pyruvate kinase M2(PKM2), lactate dehydrogenase (LDHA), glucose transporters (GLUTs), hexokinase II (HKII), phosphofructokinase-1 (PFK-1), and pyruvate dehydrogenase kinase (PDK). Here, we discuss the results of most recent studies evaluating the impact of flavonoids on HIF-1 accompanied by the regulation of critical enzymes contributing to the Warburg phenotype. Besides, flavonoid effects on glucose metabolism via regulation of HIF-1 activity represent a promising avenue in cancer-related research. At the same time, only more-in depth investigations can further elucidate the mechanistic and clinical connections between HIF-1 and cancer metabolism.
Collapse
Affiliation(s)
- Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.)
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.)
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.)
| | - Sandra Mersakova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 03601 Martin, Slovakia; (S.M.); (J.S.)
| | - Jan Strnadel
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 03601 Martin, Slovakia; (S.M.); (J.S.)
| | - Karol Kajo
- Department of Pathology, St. Elizabeth Cancer Institute Hospital, 81250 Bratislava, Slovakia;
| | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Karel Smejkal
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, Palackého třída 1946/1, 61200 Brno, Czech Republic;
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, 1477893855 Tehran, Iran;
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, 1419963114 Tehran, Iran;
| | - 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
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, 00185 Rome, Italy;
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| |
Collapse
|
28
|
Ahmed SA, Parama D, Daimari E, Girisa S, Banik K, Harsha C, Dutta U, Kunnumakkara AB. Rationalizing the therapeutic potential of apigenin against cancer. Life Sci 2020; 267:118814. [PMID: 33333052 DOI: 10.1016/j.lfs.2020.118814] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/14/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Despite the remarkable advances made in the diagnosis and treatment of cancer during the past couple of decades, it remains the second largest cause of mortality in the world, killing approximately 9.6 million people annually. The major challenges in the treatment of the advanced stage of this disease are the development of chemoresistance, severe adverse effects of the drugs, and high treatment cost. Therefore, the development of drugs that are safe, efficacious, and cost-effective remains a 'Holy Grail' in cancer research. However, the research over the past four decades shed light on the cancer-preventive and therapeutic potential of natural products and their underlying mechanism of action. Apigenin is one such compound, which is known to be safe and has significant potential in the prevention and therapy of this disease. AIM To assess the literature available on the potential of apigenin and its analogs in modulating the key molecular targets leading to the prevention and treatment of different types of cancer. METHOD A comprehensive literature search has been carried out on PubMed for obtaining information related to the sources and analogs, chemistry and biosynthesis, physicochemical properties, biological activities, bioavailability and toxicity of apigenin. KEY FINDINGS The literature search resulted in many in vitro, in vivo and a few cohort studies that evidenced the effectiveness of apigenin and its analogs in modulating important molecular targets and signaling pathways such as PI3K/AKT/mTOR, JAK/STAT, NF-κB, MAPK/ERK, Wnt/β-catenin, etc., which play a crucial role in the development and progression of cancer. In addition, apigenin was also shown to inhibit chemoresistance and radioresistance and make cancer cells sensitive to these agents. Reports have further revealed the safety of the compound and the adaptation of nanotechnological approaches for improving its bioavailability. SIGNIFICANCE Hence, the present review recapitulates the properties of apigenin and its pharmacological activities against different types of cancer, which warrant further investigation in clinical settings.
Collapse
Affiliation(s)
- Semim Akhtar Ahmed
- Cell and Molecular Biology Laboratory, Department of Zoology, Cotton University, Pan Bazar, Guwahati, Assam 781001, India
| | - Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Enush Daimari
- Cell and Molecular Biology Laboratory, Department of Zoology, Cotton University, Pan Bazar, Guwahati, Assam 781001, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Choudhary Harsha
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Uma Dutta
- Cell and Molecular Biology Laboratory, Department of Zoology, Cotton University, Pan Bazar, Guwahati, Assam 781001, India.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India.
| |
Collapse
|
29
|
Shentu Y, Jiang H, Liu X, Chen H, Yang D, Zhang J, Cheng C, Zheng Y, Zhang Y, Chen C, Zheng C, Zhou Y. Nestin Promotes Peritoneal Fibrosis by Protecting HIF1-α From Proteasomal Degradation. Front Physiol 2020; 11:517912. [PMID: 33391003 PMCID: PMC7772359 DOI: 10.3389/fphys.2020.517912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 11/20/2020] [Indexed: 12/30/2022] Open
Abstract
Background Peritoneal dialysis (PD) is a treatment for end stage renal disease patients, but it can also cause peritoneal fibrosis. Nestin is known as a neural stem cell marker and it has many functions. The hypoxia induced factor (HIF) signaling pathway can be activated under hypoxia conditions, leading to the overexpression of some angiogenesis related genes. The aim of our study is to demonstrate Nestin’s role in the development of peritoneal fibrosis (PF), and to provide a new target (Nestin) to treat PF. Methods PD mice models were constructed by an intraperitoneal administration of PDS at 10 ml/100g/d for 4 weeks. Nestin-positive cells were isolated from peritonea of Nestin-GFP mice by flow cytometry. The relationship of Nestin and HIF1-α-VEGFA pathway was detected by Nestin knockdown, Co-immunoprecipitation and immunofluorescence. Also, proteasomal activity was demonstrated by CHX and MG132 application, followed by Western blotting and Co-immunoprecipitation. Results In our experiments, we found that Nestin expression resulted in PF. Also, HIF1-α/VEGFA pathway was activated in PF. Nestin knockdown reduced the level of HIF1-α. Nestin directly bound to HIF1-α and protected HIF1-α from proteasomal degradation. Overexpression of HIF1-α reverts the fibrosis levels in Nestin-knockdown cells. In brief, Nestin inhibited the degradation of HIF1-α by mitigating its ubiquitination level, leading to the activation of HIF1-α signaling pathway, and eventually promoted PF. Conclusion We found a novel mechanism of PF that Nestin promotes by protecting HIF1-α from proteasomal degradation. Taken together, our key findings highlight a novel mechanism by which the silencing of Nestin hinders HIF1- α -induced PF.
Collapse
Affiliation(s)
- Yangping Shentu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huanchang Jiang
- Department of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyuan Liu
- Department of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hao Chen
- Department of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dicheng Yang
- Department of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinqi Zhang
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chen Cheng
- Department of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yulin Zheng
- Department of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yang Zhang
- Department of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaosheng Chen
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chenfei Zheng
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying Zhou
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
30
|
da Silva Filho AF, de Sousa LM, Consonni SR, da Rocha Pitta MG, Carvalho HF, de Melo Rêgo MJB. Galectin-3 Expression in Pancreatic Cell Lines Under Distinct Autophagy-Inducing Stimulus. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2020; 26:1187-1197. [PMID: 33107424 DOI: 10.1017/s1431927620024526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hypoxia and nutrient deprivation are responsible for inducing malignant behavior in neoplastic cells. In these conditions, metabolic stress leads the cells to enhance their autophagic flux and to activate key molecules for homeostasis maintenance. Galectin-3 (Gal-3) is upregulated in pancreatic cancer and it is activated under the hypoxic atmosphere. We aimed to analyze the most effective autophagic-inducing conditions in pancreatic ductal adenocarcinoma cells and the effect exerted under these conditions in association with hypoxia on the Gal-3 expression. Gal-3 and the microtubule-associated protein light chain 3 beta (LC3) were accessed through western blot and immunofluorescence. Degradative vacuole quantification was analyzed by transmission electronic microscopy, and inhibition of Gal-3 was performed using siRNA. According to the analyses, the most effective conditions in the inducement of autophagy for PANC-1 and MIA PaCa-2 cells were nutritional deprivation and complete amino acid/glucose deprivation, respectively. PANC-1 cells presented higher Gal-3 when they were submitted to 24 h of nutritional deprivation alone and simultaneously nutritional and oxygen deprivation. Inhibition of Gal-3 causes a decrease of LC3 levels in all experimental conditions. These results confirm that Gal-3 is modulated by microenvironment factors and the possibility of Gal-3 participating in an adaptive response from PDAC cells to extreme conditions.
Collapse
Affiliation(s)
- Antônio Felix da Silva Filho
- Immunomodulation and New Therapy Approaches Laboratory (LINAT), Biochemistry Department, Federal University of Pernambuco (UFPE), Cidade Universitária, Recife, Pernambuco50670-901, Brazil
| | - Lizandra Maia de Sousa
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, São Paulo13083-970, Brazil
| | - Silvio Roberto Consonni
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, São Paulo13083-970, Brazil
| | - Maira Galdino da Rocha Pitta
- Immunomodulation and New Therapy Approaches Laboratory (LINAT), Biochemistry Department, Federal University of Pernambuco (UFPE), Cidade Universitária, Recife, Pernambuco50670-901, Brazil
| | - Hernandes Faustino Carvalho
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, São Paulo13083-970, Brazil
| | - Moacyr Jesus Barreto de Melo Rêgo
- Immunomodulation and New Therapy Approaches Laboratory (LINAT), Biochemistry Department, Federal University of Pernambuco (UFPE), Cidade Universitária, Recife, Pernambuco50670-901, Brazil
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Therapeutic Innovation Research Center- Suelly Galdino (NUPIT-SG), Biochemistry Department, Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco50670-901, Brazil
| |
Collapse
|
31
|
Ashrafizadeh M, Bakhoda MR, Bahmanpour Z, Ilkhani K, Zarrabi A, Makvandi P, Khan H, Mazaheri S, Darvish M, Mirzaei H. Apigenin as Tumor Suppressor in Cancers: Biotherapeutic Activity, Nanodelivery, and Mechanisms With Emphasis on Pancreatic Cancer. Front Chem 2020; 8:829. [PMID: 33195038 PMCID: PMC7593821 DOI: 10.3389/fchem.2020.00829] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is the most lethal malignancy of the gastrointestinal tract. Due to its propensity for early local and distant spread, affected patients possess extremely poor prognosis. Currently applied treatments are not effective enough to eradicate all cancer cells, and minimize their migration. Besides, these treatments are associated with adverse effects on normal cells and organs. These therapies are not able to increase the overall survival rate of patients; hence, finding novel adjuvants or alternatives is so essential. Up to now, medicinal herbs were utilized for therapeutic goals. Herbal-based medicine, as traditional biotherapeutics, were employed for cancer treatment. Of them, apigenin, as a bioactive flavonoid that possesses numerous biological properties (e.g., anti-inflammatory and anti-oxidant effects), has shown substantial anticancer activity. It seems that apigenin is capable of suppressing the proliferation of cancer cells via the induction of cell cycle arrest and apoptosis. Besides, apigenin inhibits metastasis via down-regulation of matrix metalloproteinases and the Akt signaling pathway. In pancreatic cancer cells, apigenin sensitizes cells in chemotherapy, and affects molecular pathways such as the hypoxia inducible factor (HIF), vascular endothelial growth factor (VEGF), and glucose transporter-1 (GLUT-1). Herein, the biotherapeutic activity of apigenin and its mechanisms toward cancer cells are presented in the current review to shed some light on anti-tumor activity of apigenin in different cancers, with an emphasis on pancreatic cancer.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Mohammad Reza Bakhoda
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Bahmanpour
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khandan Ilkhani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Pooyan Makvandi
- Centre for Micro-BioRobotics, Istituto Italiano di Tecnologia, Pisa, Italy.,Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Samaneh Mazaheri
- Department of Analytical Chemistry, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Maryam Darvish
- Department of Medical Biotechnology, Faculty of Medicine, Arak University of Medical Science, Arak, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
32
|
Flavones and flavonols may have clinical potential as CK2 inhibitors in cancer therapy. Med Hypotheses 2020; 141:109723. [DOI: 10.1016/j.mehy.2020.109723] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/27/2020] [Accepted: 04/08/2020] [Indexed: 01/16/2023]
|
33
|
Thyagarajan A, Forino AS, Konger RL, Sahu RP. Dietary Polyphenols in Cancer Chemoprevention: Implications in Pancreatic Cancer. Antioxidants (Basel) 2020; 9:antiox9080651. [PMID: 32717779 PMCID: PMC7464582 DOI: 10.3390/antiox9080651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
Naturally occurring dietary agents present in a wide variety of plant products, are rich sources of phytochemicals possessing medicinal properties, and thus, have been used in folk medicine for ages to treat various ailments. The beneficial effects of such dietary components are frequently attributed to their anti-inflammatory and antioxidant properties, particularly in regards to their antineoplastic activities. As many tumor types exhibit greater oxidative stress levels that are implicated in favoring autonomous cell growth activation, most chemotherapeutic agents can also enhance tumoral oxidative stress levels in part via generating reactive oxygen species (ROS). While ROS-mediated imbalance of the cellular redox potential can provide novel drug targets, as a consequence, this ROS-mediated excessive damage to cellular functions, including oncogenic mutagenesis, has also been implicated in inducing chemoresistance. This remains one of the major challenges in the treatment and management of human malignancies. Antioxidant-enriched natural compounds offer one of the promising approaches in mitigating some of the underlying mechanisms involved in tumorigenesis and metastasis, and therefore, have been extensively explored in cancer chemoprevention. Among various groups of dietary phytochemicals, polyphenols have been extensively explored for their underlying chemopreventive mechanisms in other cancer models. Thus, the current review highlights the significance and mechanisms of some of the highly studied polyphenolic compounds, with greater emphasis on pancreatic cancer chemoprevention.
Collapse
Affiliation(s)
- Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of medicine Wright State University, Dayton, OH 45435, USA
- Correspondence: (A.T.); (R.P.S.); Tel.: +1-937-775-4603 (R.P.S.)
| | - Andrew S. Forino
- Department of Anatomy and Physiology, Boonshoft School of medicine Wright State University, Dayton, OH 45435, USA;
| | - Raymond L. Konger
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Ravi P. Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of medicine Wright State University, Dayton, OH 45435, USA
- Correspondence: (A.T.); (R.P.S.); Tel.: +1-937-775-4603 (R.P.S.)
| |
Collapse
|
34
|
Yao L, Fan Z, Han S, Sun N, Che H. Apigenin Attenuates the Allergic Reactions by Competitively Binding to ER With Estradiol. Front Pharmacol 2020; 11:1046. [PMID: 32765268 PMCID: PMC7378534 DOI: 10.3389/fphar.2020.01046] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Apigenin (API) is a natural phytoestrogen with properties including anti-inflammatory and other abilities. This study aims to 1) systematically validate that excessive estrogen exacerbates allergic reactions; 2) explore the anti-allergic effects and mechanisms of API. We conduct a survey of college students, indicating that of the 505 effective results, 70 individuals were self-reported allergic and 74.1% of them were women, which proved the gender difference in allergic reactions. BALB/c mice are grouped into the negative control group (N-Ctrl), the OVA-sensitized group (P-Ctrl), the estrogenized OVA-sensitized group (E2), and three treatment groups administrating different dose of API (E2 + API/L/M/H). In vivo data indicated that API treatment significantly inhibited the enhancement of estradiol on clinical symptoms. Moreover, we found that high doses of API inhibited Th2 type humoral response and mast cell degranulation levels in vivo and in vitro. Additionally, medium, and high doses of API significantly reduced the potentiation of estradiol on ER expression, attenuated the transmission of estrogen/ER signaling, thereby inhibiting the phosphorylation of ERK1/2 and JNK1/2/3 in the MAPK. Besides, we found that API competitively bound to ER with estradiol, and showed a weak selectivity to ERβ. Overall, we identified API can be beneficial in allergic disease.
Collapse
Affiliation(s)
- Lu Yao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zhuoyan Fan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Shiwen Han
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Na Sun
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Huilian Che
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
35
|
Xi Y, Yuan P, Li T, Zhang M, Liu MF, Li B. hENT1 reverses chemoresistance by regulating glycolysis in pancreatic cancer. Cancer Lett 2020; 479:112-122. [PMID: 32200037 DOI: 10.1016/j.canlet.2020.03.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/10/2020] [Accepted: 03/16/2020] [Indexed: 12/13/2022]
Abstract
Gemcitabine (GEM) chemotherapy, as the first-line regimen for pancreatic cancer, tends to induce drug resistance, which ultimately worsens the prognosis of patients with pancreatic cancer. Our previous study indicated a close correlation between pancreatic cancer progression and glucose metabolism, especially at the chemoresistant stage, highlighting the importance of the application of 18F-FDG PET dual-phase imaging in the early detection of pancreatic cancer. We speculate that glycolysis, participates in the development of chemoresistance in pancreatic cancer. In this article, we wanted to determine whether manipulating hENT1 expression in pancreatic cancer cells can reverse GEM chemoresistance and whether glucose transport and glycolysis are involved during this process. We found that hENT1 reversed GEM-induced drug resistance by inhibiting glycolysis and altering glucose transport mediated by HIF-1α in pancreatic cancer. Our findings also suggest that 18F-FDG PET dual-phase imaging after the 4th chemotherapy treatment can accurately identify drug-resistant pancreatic tumors and improve hENT1 reversal therapy. Our findings highlight that the dynamic observation of (retention index) RI changes from the beginning of treatment can also be helpful for evaluating the therapeutic effect.
Collapse
Affiliation(s)
- Yun Xi
- Department of Nuclear Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, 197 Rui Jin 2(nd) Road, Shanghai, 200025, China
| | - Peng Yuan
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, 320 Yueyang Road, Shanghai, 200031, China
| | - Ting Li
- Department of Nuclear Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, 197 Rui Jin 2(nd) Road, Shanghai, 200025, China
| | - Min Zhang
- Department of Nuclear Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, 197 Rui Jin 2(nd) Road, Shanghai, 200025, China
| | - Mo-Fang Liu
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, 320 Yueyang Road, Shanghai, 200031, China; Center for Excellence in Molecular Cell Science 8 School of Life Science and Technology, Shanghai Tech University, 393 Middle Huaxia Road, Shanghai, 201210, China; Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai, 200438, China
| | - Biao Li
- Department of Nuclear Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, 197 Rui Jin 2(nd) Road, Shanghai, 200025, China.
| |
Collapse
|
36
|
Imran M, Aslam Gondal T, Atif M, Shahbaz M, Batool Qaisarani T, Hanif Mughal M, Salehi B, Martorell M, Sharifi-Rad J. Apigenin as an anticancer agent. Phytother Res 2020; 34:1812-1828. [PMID: 32059077 DOI: 10.1002/ptr.6647] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/03/2020] [Accepted: 01/31/2020] [Indexed: 12/26/2022]
Abstract
Apigenin is an edible plant-derived flavonoid that has been reported as an anticancer agent in several experimental and biological studies. It exhibits cell growth arrest and apoptosis in different types of tumors such as breast, lung, liver, skin, blood, colon, prostate, pancreatic, cervical, oral, and stomach, by modulating several signaling pathways. Apigenin induces apoptosis by the activation of extrinsic caspase-dependent pathway by upregulating the mRNA expressions of caspase-3, caspase-8, and TNF-α. It induces intrinsic apoptosis pathway as evidenced by the induction of cytochrome c, Bax, and caspase-3, while caspase-8, TNF-α, and B-cell lymphoma 2 levels remained unchanged in human prostate cancer PC-3 cells. Apigenin treatment leads to significant downregulation of matrix metallopeptidases-2, -9, Snail, and Slug, suppressing invasion. The expressions of NF-κB p105/p50, PI3K, Akt, and the phosphorylation of p-Akt decreases after treatment with apigenin. However, apigenin-mediated treatment significantly reduces pluripotency marker Oct3/4 protein expression which might be associated with the downregulation of PI3K/Akt/NF-κB signaling.
Collapse
Affiliation(s)
- Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore-Lahore, Lahore, Pakistan
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Deakin University, Melbourne, Victoria, Australia
| | - Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Muhammad Shahbaz
- Department of Food Science and Technology, MNS-University of Agriculture Multan, Multan, Pakistan
| | - Tahira Batool Qaisarani
- Department of Agricultural Engineering and Technology, Ghazi University, Dera Ghazi Khan, Pakistan
| | - Muhammad Hanif Mughal
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore-Lahore, Lahore, Pakistan
| | - Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción, Chile.,Unidad de Desarrollo Tecnológico, UDT, Universidad de Concepción, Concepción, Chile
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Deng X, Peng Y, Zhao J, Lei X, Zheng X, Xie Z, Tang G. Anticancer Activity of Natural Flavonoids: Inhibition of HIF-1α Signaling Pathway. CURR ORG CHEM 2020. [DOI: 10.2174/1385272823666191203122030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rapid tumor growth is dependent on the capability of tumor blood vessels and
glycolysis to provide oxygen and nutrients. Tumor hypoxia is a common characteristic of
many solid tumors, and it essentially happens when the growth of the tumor exceeds the
concomitant angiogenesis. Hypoxia-inducible factor 1 (HIF-1) as the critical transcription
factor in hypoxia regulation is activated to adapt to this hypoxia situation. Flavonoids,
widely distributed in plants, comprise many polyphenolic secondary metabolites, possessing
broadspectrum pharmacological activities, including their potentiality as anticancer
agents. Due to their low toxicity, intense efforts have been made for investigating natural
flavonoids and their derivatives that can be used as HIF-1α inhibitors for cancer therapy
during the past few decades. In this review, we sum up the findings concerning the inhibition
of HIF-1α by natural flavonoids in the last few years and propose the idea of designing tumor vascular and
glycolytic multi-target inhibitors with HIF-1α as one of the targets.
Collapse
Affiliation(s)
- Xiangping Deng
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Yijiao Peng
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Jingduo Zhao
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xiaoyong Lei
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xing Zheng
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zhizhong Xie
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
38
|
Zhang X, Wang J, Wu J, Jiang X, Pei X, Chen J, Wan Q, Huang C. Dimethyloxalylglycine improves angiogenesis of ZIF-8-coated implant. J Biomater Appl 2019; 34:396-407. [PMID: 31117856 DOI: 10.1177/0885328219850976] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Xin Zhang
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Wang
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,2 Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiaxin Wu
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoge Jiang
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xibo Pei
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,2 Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junyu Chen
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,2 Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianbing Wan
- 1 State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,2 Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chao Huang
- 3 College of Chemistry, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
A critical review on anti-angiogenic property of phytochemicals. J Nutr Biochem 2019; 71:1-15. [PMID: 31174052 DOI: 10.1016/j.jnutbio.2019.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/12/2019] [Accepted: 04/25/2019] [Indexed: 12/14/2022]
Abstract
Angiogenesis, a process involved in neovascularization, has been found to be associated with several metabolic diseases like cancer, retinopathy etc. Thus, currently, the focus on anti-angiogenic therapy for treatment and prevention of diseases has gained significant attention. Currently available Food and Drug Administration (FDA) approved drugs are targeting either vascular endothelial growth factor or it's receptor, but in the long term, these approaches were shown to cause several side effects and the chances of developing resistance to these drugs is also high. Therefore, identification of safe and cost-effective anti-angiogenic molecules is highly imperative. Over the past decades, dietary based natural compounds have been studied for their anti-angiogenic potential which provided avenues in improving the angiogenesis based therapy. In this review, major emphasis is given to the molecular mechanism behind anti-angiogenic effect of natural compounds from dietary sources.
Collapse
|
40
|
Sliker BH, Goetz BT, Peters HL, Poelaert BJ, Borgstahl GEO, Solheim JC. Beta 2-microglobulin regulates amyloid precursor-like protein 2 expression and the migration of pancreatic cancer cells. Cancer Biol Ther 2019; 20:931-940. [PMID: 30810435 DOI: 10.1080/15384047.2019.1580414] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Beta 2-microglobulin (β2m) is a component of the major histocompatibility complex (MHC) class I molecule, which presents tumor antigens to T lymphocytes to trigger cancer cell destruction. Notably, β2m has been reported as persistently expressed, rather than down regulated, in some tumor types. For renal cell and oral squamous cell carcinomas, β2m expression has been linked to increased migratory capabilities. The migratory ability of pancreatic cancer cells contributes to their metastatic tendencies and lethal nature. Therefore, in this study, we examined the impact of β2m on pancreatic cancer cell migration. We found that β2m protein is amply expressed in several human pancreatic cancer cell lines (S2-013, PANC-1, and MIA PaCa-2). Reducing β2m expression by short interfering RNA (siRNA) transfection significantly slowed the migration of the PANC-1 and S2-013 cancer cell lines, but increased the migration of the MIA PaCa-2 cell line. The amyloid precursor-like protein 2 (APLP2) has been documented as contributing to pancreatic cancer cell migration, invasiveness, and metastasis. We have previously shown that β2m/HLA class I/peptide complexes associate with APLP2 in S2-013 cells, and in this study we also detected their association in PANC-1 cells but not MIA PaCa-2 cells. In addition, siRNA down regulation of β2m expression diminished the expression of APLP2 in S2-013 and PANC-1 but heightened the level of APLP2 in MIA PaCa-2 cells, consistent with our migration data and co-immunoprecipitation data. Thus, our findings indicate that β2m regulates pancreatic cancer cell migration, and furthermore suggest that APLP2 is an intermediary in this process.
Collapse
Affiliation(s)
- Bailee H Sliker
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA
| | - Benjamin T Goetz
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA
| | - Haley L Peters
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA
| | - Brittany J Poelaert
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA
| | - Gloria E O Borgstahl
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA.,c Department of Biochemistry and Molecular Biology , University of Nebraska Medical Center , Omaha , NE , USA.,d Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , NE , USA
| | - Joyce C Solheim
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA.,c Department of Biochemistry and Molecular Biology , University of Nebraska Medical Center , Omaha , NE , USA.,e Department of Pathology and Microbiology , University of Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
41
|
Kurahara H, Maemura K, Mataki Y, Sakoda M, Iino S, Kawasaki Y, Arigami T, Mori S, Kijima Y, Ueno S, Shinchi H, Natsugoe S. Significance of 18F-Fluorodeoxyglucose (FDG) Uptake in Response to Chemoradiotherapy for Pancreatic Cancer. Ann Surg Oncol 2018; 26:644-651. [PMID: 30523468 DOI: 10.1245/s10434-018-07098-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND A metabolic shift to glycolysis is reportedly involved in radioresistance. We examined whether pretreatment 18F-fluorodeoxyglucose positron emission tomography (FDG-PET), which can detect enhanced glucose uptake, was able to predict the therapeutic response to chemoradiotherapy (CRT) in patients with pancreatic cancer (PC). METHODS Of 125 PC patients (75 unresectable and 50 borderline resectable), 37 and 26 underwent induction chemotherapy before CRT and surgical resection after CRT, respectively. FDG-PET was performed at three different institutions. RESULTS Of the 88 patients who underwent upfront CRT, 31 (35%), 34 (39%), and 23 (26%) showed a partial response (PR), stable disease, and progressive disease, respectively. The tumor PR rate was an independent factor associated with longer overall survival (OS) on multivariate analysis. We evaluated the optimal cut-off of maximum standardized uptake values (SUVmax) at initial diagnosis to detect the tumor PR rate at the three institutions separately. The SUVmax was independently associated with tumor response rate on multivariate analysis. In the low SUVmax group, induction chemotherapy had no significant impact on OS. In contrast, induction chemotherapy was significantly associated with longer OS in the high SUVmax group. CONCLUSIONS FDG-PET SUVmax was significantly associated with the therapeutic response to CRT in PC patients. Moreover, induction chemotherapy may improve the prognosis of patients with a high SUVmax tumor.
Collapse
Affiliation(s)
- Hiroshi Kurahara
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan.
| | - Kosei Maemura
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Yuko Mataki
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Masahiko Sakoda
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Satoshi Iino
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Yota Kawasaki
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Takaaki Arigami
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Shinichiro Mori
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Yuko Kijima
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Shinichi Ueno
- Clinical Oncology, Kagoshima University, Kagoshima, Japan
| | | | - Shoji Natsugoe
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
42
|
Kashyap D, Sharma A, Tuli HS, Sak K, Garg VK, Buttar HS, Setzer WN, Sethi G. Apigenin: A natural bioactive flavone-type molecule with promising therapeutic function. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.037] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
43
|
An Array of Bioactive Compounds From Australian Eucalypts and Their Relevance in Pancreatic Cancer Therapeutics. Pancreas 2018; 47:690-707. [PMID: 29894418 DOI: 10.1097/mpa.0000000000001074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer (PC) is one of the most devastating human cancers, and despite the significant advances in the current therapeutic options, the overall survival rate for PC has remained static for the past 50 years. Plant-derived bioactive compounds play a vital role in cancer therapeutics by providing new lead compounds for future drug development. Therefore, the isolation, characterization, and identification of new bioactive compounds for the prevention and treatment of cancer continue to be an important aspect of natural product research. Many in vitro and in vivo studies published in the last few decades have established strong links between the phytochemical profile of eucalypts and anticancer activity. However, only a small number of these reports have attempted to demonstrate a relationship between the biological activity of eucalypt extracts and PC. This review focuses on potential anti-PC effects of an array of bioactive compounds present in various species of eucalypts. It also highlights the necessity for further in vitro and in vivo studies to develop a complete understanding of the potential this group of plants has for the development of potent and specific chemotherapeutic drugs for PC.
Collapse
|
44
|
Bhuyan DJ, Vuong QV, Bond DR, Chalmers AC, Bowyer MC, Scarlett CJ. Eucalyptus microcorys leaf extract derived HPLC-fraction reduces the viability of MIA PaCa-2 cells by inducing apoptosis and arresting cell cycle. Biomed Pharmacother 2018; 105:449-460. [PMID: 29879629 DOI: 10.1016/j.biopha.2018.05.150] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 12/22/2022] Open
Abstract
New therapeutic strategies such as the development of novel drugs and combinatorial therapies with existing chemotherapeutic agents are urgently needed to improve the clinical prognosis of pancreatic cancer. We have previously reported the antiproliferative properties of aqueous crude Eucalyptus microcorys extract against pancreatic cancer cell lines. In this study, bioassay-guided fractionation of the aqueous crude E. microcorys extract using RP-HPLC and subsequent assessment of the resultant fractions (F1-F5) for their antioxidant activity and cytotoxicity against pancreatic cancer cell lines were performed. The molecular mechanisms associated with the cytotoxicity was characterised by studying the effects of the most potent fraction-1 (F1) on apoptosis and cell cycle profiles as well as its phytochemical constituents by LC-ESI/MS/MS. F1 displayed significantly greater antioxidant activity in three different assays (p < 0.05). Moreover, F1 exhibited significantly greater antiproliferative activity (IC50 = 93.11 ± 3.43 μg/mL) against MIA PaCa-2 cells compared to the other four fractions (p < 0.05). F1 induced apoptosis by regulating key apoptotic proteins- Bcl-2, Bak, Bax, cleaved PARP, procaspase-3 and cleaved caspase-3 in MIA PaCa-2 cells, suggesting the involvement of intrinsic mitochondrial apoptotic pathway and arrested cells at G2/M phase. A combination of gemcitabine and F1 exerted a greater effect on apoptosis and cell cycle arrest than F1 or gemcitabine alone (p < 0.05). LC-ESI/MS/MS revealed the tentative identities of phytochemicals present in F1 and their similarities with the phenolic compounds previously reported in Eucalyptus with antipancreatic cancer activity. Our study shows that the polyphenol and antioxidant-rich fraction of E. microcorys extract is a promising candidate for developing mono or combination therapies against pancreatic cancer.
Collapse
Affiliation(s)
- Deep Jyoti Bhuyan
- Pancreatic Cancer Research Group, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia; School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia.
| | - Quan V Vuong
- Pancreatic Cancer Research Group, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia; School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia
| | - Danielle R Bond
- Pancreatic Cancer Research Group, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia; School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia
| | - Anita C Chalmers
- School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia
| | - Michael C Bowyer
- Pancreatic Cancer Research Group, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia; School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia
| | - Christopher J Scarlett
- Pancreatic Cancer Research Group, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia; School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia
| |
Collapse
|
45
|
Tang L, Wei F, Wu Y, He Y, Shi L, Xiong F, Gong Z, Guo C, Li X, Deng H, Cao K, Zhou M, Xiang B, Li X, Li Y, Li G, Xiong W, Zeng Z. Role of metabolism in cancer cell radioresistance and radiosensitization methods. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:87. [PMID: 29688867 PMCID: PMC5914062 DOI: 10.1186/s13046-018-0758-7] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Radioresistance is a major factor leading to the failure of radiotherapy and poor prognosis in tumor patients. Following the application of radiotherapy, the activity of various metabolic pathways considerably changes, which may result in the development of resistance to radiation. MAIN BODY Here, we discussed the relationships between radioresistance and mitochondrial and glucose metabolic pathways, aiming to elucidate the interplay between the tumor cell metabolism and radiotherapy resistance. In this review, we additionally summarized the potential therapeutic targets in the metabolic pathways. SHORT CONCLUSION The aim of this review was to provide a theoretical basis and relevant references, which may lead to the improvement of the sensitivity of radiotherapy and prolong the survival of cancer patients.
Collapse
Affiliation(s)
- Le Tang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Wei
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yingfen Wu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yi He
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lei Shi
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Can Guo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Deng
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ke Cao
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bo Xiang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
46
|
Park K, Lee HE, Lee SH, Lee D, Lee T, Lee YM. Molecular and functional evaluation of a novel HIF inhibitor, benzopyranyl 1,2,3-triazole compound. Oncotarget 2018; 8:7801-7813. [PMID: 27999195 PMCID: PMC5352362 DOI: 10.18632/oncotarget.13955] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022] Open
Abstract
Hypoxia occurs in a variety of pathological events, including the formation of solid tumors. Hypoxia-inducible factor (HIF)-1α is stabilized under hypoxic conditions and is a key molecule in tumor growth and angiogenesis. Seeking to develop novel cancer therapeutics, we investigated small molecules from our in-house chemical libraries to target HIF-1α. We employed a dual-luciferase assay that uses a luciferase (Luc) reporter vector harboring five copies of hypoxia-responsive element (HRE) in the promoter. Under hypoxic conditions that increased Luc reporter activity by four-fold, we screened 144 different compounds, nine of which showed 30–50% inhibition of hypoxia-induced Luc reporter activity. Among these, “Compound 12, a benzopyranyl 1,2,3-triazole” was the most efficient at inhibiting the expression of HIF-1α under hypoxic conditions, reducing its expression by 80%. Under hypoxic conditions, the half maximal IC50 of the compound was 24 nM in HEK-293 human embryonic kidney cells, and 2 nM in A549 human lung carcinoma cells. Under hypoxic conditions, Compound 12 increased hydroxylated HIF-1α levels and HIF-1α ubiquitination, and also dose-dependently decreased HIF-1α target gene expression as well as vascular endothelial growth factor (VEGF) secretion. Furthermore, this compound inhibited VEGF-induced in vitro angiogenesis in human umbilical vein endothelial cells (HUVECs), and in vivo, it inhibited chick chorioallantoic membrane angiogenesis. In allogaft assays, cotreatment with Compound 12 and gefitinib significantly inhibited tumor growth and angiogenesis. Compound 12 can be a novel inhibitor of HIF-1α by accelerating its degradation, and shows much potential as an anti-cancer agent through its ability to suppress tumor growth and angiogenesis.
Collapse
Affiliation(s)
- Kyunghye Park
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, National Basic Research Laboratory of Vascular Homeostasis Regulation, Kyungpook National University, Buk-gu, 702-701, Daegu, Republic of Korea
| | - Hye Eun Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, National Basic Research Laboratory of Vascular Homeostasis Regulation, Kyungpook National University, Buk-gu, 702-701, Daegu, Republic of Korea
| | - Sun Hee Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, National Basic Research Laboratory of Vascular Homeostasis Regulation, Kyungpook National University, Buk-gu, 702-701, Daegu, Republic of Korea
| | - Doohyun Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Buk-gu, 702-701, Daegu, Republic of Korea
| | - Taeho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Buk-gu, 702-701, Daegu, Republic of Korea
| | - You Mie Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, National Basic Research Laboratory of Vascular Homeostasis Regulation, Kyungpook National University, Buk-gu, 702-701, Daegu, Republic of Korea.,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Buk-gu, 702-701, Daegu, Republic of Korea
| |
Collapse
|
47
|
Kurahara H, Maemura K, Mataki Y, Sakoda M, Iino S, Kawasaki Y, Arigami T, Mori S, Kijima Y, Ueno S, Shinchi H, Natsugoe S. Significance of Glucose Transporter Type 1 (GLUT-1) Expression in the Therapeutic Strategy for Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol 2018; 25:1432-1439. [PMID: 29404819 DOI: 10.1245/s10434-018-6357-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND This study aimed to examine the prognostic relevance of glucose transporter type 1 (GLUT-1), which is a key regulator of the glucose metabolism. In particular, the study aimed to examine the association between GLUT-1 expression and the therapeutic effect of chemoradiotherapy (CRT) in pancreatic ductal adenocarcinoma (PDAC). METHODS Patients with PDAC were enrolled in the study. Patients with distant metastases and those who received only chemotherapy as treatment were excluded from the study. Specimens for immunohistochemical evaluations were obtained through surgical resection and endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) of the primary tumor before any treatment. RESULTS This study included 197 patients. Of these 197 patients, 100 underwent upfront surgery, and 97 received neoadjuvant CRT (NACRT), which was performed mainly for patients with locally advanced tumors. Of the 97 patients who received NACRT, 21 later underwent surgical resection. For the patients who underwent upfront surgery, low GLUT-1 expression was an independent factor for a better prognosis. For the patients who underwent NACRT, low GLUT-1 expression was significantly associated with greater tumor size reduction, a higher resection rate, and a better prognosis. Additionally, GLUT-1 expression was significantly increased after NACRT treatment. CONCLUSIONS Among the patients with PDAC, those with low GLUT-1 expression in the primary tumor had a better prognosis those with high GLUT-1 expression. Moreover, the patients with low GLUT-1 expression displayed a better therapeutic response to NACRT.
Collapse
Affiliation(s)
- Hiroshi Kurahara
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan.
| | - Kosei Maemura
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Yuko Mataki
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Masahiko Sakoda
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Satoshi Iino
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Yota Kawasaki
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Takaaki Arigami
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Shinichiro Mori
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Yuko Kijima
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| | - Shinichi Ueno
- Department of Clinical Oncology, Kagoshima University, Kagoshima, Japan
| | - Hiroyuki Shinchi
- Department of Health Sciences, Kagoshima University, Kagoshima, Japan
| | - Shoji Natsugoe
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
48
|
Shankar E, Goel A, Gupta K, Gupta S. Plant flavone apigenin: An emerging anticancer agent. CURRENT PHARMACOLOGY REPORTS 2017; 3:423-446. [PMID: 29399439 PMCID: PMC5791748 DOI: 10.1007/s40495-017-0113-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Research in cancer chemoprevention provides convincing evidence that increased intake of vegetables and fruits may reduce the risk of several human malignancies. Phytochemicals present therein provide beneficial anti-inflammatory and antioxidant properties that serve to improve the cellular microenvironment. Compounds known as flavonoids categorized anthocyanidins, flavonols, flavanones, flavonols, flavones, and isoflavones have shown considerable promise as chemopreventive agents. Apigenin (4', 5, 7-trihydroxyflavone), a major plant flavone, possessing antioxidant, anti-inflammatory, and anticancer properties affecting several molecular and cellular targets used to treat various human diseases. Epidemiologic and case-control studies have suggested apigenin reduces the risk of certain cancers. Studies demonstrate that apigenin retain potent therapeutic properties alone and/or increases the efficacy of several chemotherapeutic drugs in combination on a variety of human cancers. Apigenin's anticancer effects could also be due to its differential effects in causing minimal toxicity to normal cells with delayed plasma clearance and slow decomposition in liver increasing the systemic bioavailability in pharmacokinetic studies. Here we discuss the anticancer role of apigenin highlighting its potential activity as a chemopreventive and therapeutic agent. We also highlight the current caveats that preclude apigenin for its use in the human trials.
Collapse
Affiliation(s)
- Eswar Shankar
- Department of Urology, The James and Eilleen Dicke Laboratory, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Urology, The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Aditi Goel
- Department of Biology, School of Undergraduate Studies, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Karishma Gupta
- Department of Urology, The James and Eilleen Dicke Laboratory, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Urology, The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Sanjay Gupta
- Department of Urology, The James and Eilleen Dicke Laboratory, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Urology, The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA
- Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
49
|
Fan J, Mei J, Zhang MZ, Yuan F, Li SZ, Yu GR, Chen LH, Tang Q, Xian CJ. Clinicopathological significance of glucose transporter protein-1 overexpression in human osteosarcoma. Oncol Lett 2017; 14:2439-2445. [PMID: 28781680 DOI: 10.3892/ol.2017.6437] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 04/13/2017] [Indexed: 01/09/2023] Open
Abstract
Although previous studies have demonstrated that Glut-1 is the predominant glucose transporter, is significantly overexpressed in various types of tumor and is correlated with poor prognosis, the potential function and clinical value of Glut-1 expression in osteosarcoma remains largely unclear. In particular, the prospective associations between Glut-1 expression levels and clinicopathological factors remains to be elucidated. In the present study, immunohistochemistry was performed to detect Glut-1 protein expression in 51 paired osteosarcoma specimens and adjacent non-cancerous tissues, and reverse transcription-quantitative polymerase chain reaction analysis was performed to examine Glut-1 mRNA expression levels in 6 pairs of these tissues. Statistical analyses were conducted to determine the associations between Glut-1 expression and various clinicopathological parameters. Glut-1 protein was revealed to be overexpressed in 38 (74.5%) osteosarcoma tissues, but only in 6 (11.8%) adjacent non-cancerous tissues. Glut-1 mRNA levels were also upregulated in osteosarcoma tissues compared with adjacent non-cancerous tissues. While there were no clear statistical relationships between Glut-1 expression and patient sex, resection, tumor location, size, T stage and adjuvant treatment, Glut-1 expression levels were significantly associated with age, tumor-node-metastasis stage, lymph node metastasis and survival. The median survival time in patients with low Glut-1 expression levels was longer than in patients with a high expression level. Glut-1 was significantly overexpressed in osteosarcoma tissues, and Glut-1 expression was associated with clinicopathological factors which upregulate the invasion and metastasis of osteosarcoma, and may be a potential predictor of survival in patients with osteosarcoma.
Collapse
Affiliation(s)
- Jian Fan
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Jiong Mei
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Ming-Zhu Zhang
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Feng Yuan
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Shan-Zhu Li
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Guang-Rong Yu
- Department of Orthopedics, Tongji Hospital, Tongji University, Shanghai 200065, P.R. China
| | - Long-Hui Chen
- Pi-wei Institute, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Qian Tang
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| |
Collapse
|
50
|
Wang G, Wang JJ, Guan R, Du L, Gao J, Fu XL. Strategies to Target Glucose Metabolism in Tumor Microenvironment on Cancer by Flavonoids. Nutr Cancer 2017; 69:534-554. [PMID: 28323500 DOI: 10.1080/01635581.2017.1295090] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The imbalance between glucose metabolism and cancer cell growth in tumor microenvironment (TME), which are closely related with the occurrence and progression of cancer. Accumulating evidence has demonstrated that flavonoids exert many biological properties, including antioxidant and anticarcinogenic activities. Recently, the roles and applications of flavonoids, particularly in relation to glucose metabolism in cancers, have been highlighted. Thus, the identification of flavonoids targeting alternative glucose metabolism pathways in TME may represent an attractive approach to the more effective therapeutic strategies for cancer. In this review, we will focus on the roles of flavonoids in regulating glucose metabolism and cancer cell growth in TME, such as proliferation advantage, cell mobility, and chemoresistance to cancer, as well as modifiers of thermal sensitivity. Not only have such large-scale endeavors been useful in providing fundamental insights into natural and synthesized flavonoids that can prevent and treat cancer, but also have led to the discovery of potential targets for cancer therapy.
Collapse
Affiliation(s)
- Gang Wang
- a Department of Pharmaceutics , Jiangsu University , Shanghai , China
- b Hubei University of Medicine , Shiyan , China
| | - Jun-Jie Wang
- a Department of Pharmaceutics , Jiangsu University , Shanghai , China
- b Hubei University of Medicine , Shiyan , China
| | - Rui Guan
- b Hubei University of Medicine , Shiyan , China
| | - Li Du
- a Department of Pharmaceutics , Jiangsu University , Shanghai , China
| | - Jing Gao
- c Jiangsu University Health Science Center , Jiangsu , China
| | - Xing-Li Fu
- c Jiangsu University Health Science Center , Jiangsu , China
| |
Collapse
|