1
|
Zhang ZS, Gao ZX, He JJ, Ma C, Tao HT, Zhu FY, Cheng YN, Xie CQ, Li JQ, Liu ZZ, Hou LL, Sun H, Xie SQ, Fang D. Andrographolide sensitizes glioma to temozolomide by inhibiting DKK1 expression. Br J Cancer 2024; 131:1387-1398. [PMID: 39266624 PMCID: PMC11473956 DOI: 10.1038/s41416-024-02842-0] [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: 07/04/2023] [Revised: 08/17/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Temozolomide (TMZ) is the first-line chemotherapeutic drug for gliomas treatment. However, the clinical efficacy of TMZ in glioma patients was very limited. Therefore, it is urgently needed to discover a novel approach to increase the sensitivity of glioma cells to TMZ. METHODS Western blot, immunohistochemical staining, and qRT-PCR assays were used to explore the mechanisms underlying TMZ promoting DKK1 expression and andrographolide (AND) inhibiting DKK1 expression. HPLC was used to detect the ability of andrographolide (AND) to penetrate the blood-brain barrier. MTT assay, bioluminescence images, magnetic resonance imaging (MRI) and H&E staining were employed to measure the proliferative activity of glioma cells and the growth of intracranial tumors. RESULTS TMZ can promote DKK1 expression in glioma cells and brain tumors of an orthotopic model of glioma. DKK1 could promote glioma cell proliferation and tumor growth in an orthotopic model of glioma. Mechanistically, TMZ increased EGFR expression and subsequently induced the activation of its downstream MEK-ERK and PI3K-Akt pathways, thereby promoting DKK1 expression in glioma cells. Andrographolide inhibited TMZ-induced DKK1 expression through inactivating MEK-ERK and PI3K-Akt pathways. Andrographolide can cross the blood-brain barrier, the combination of TMZ and andrographolide not only improved the anti-tumor effects of TMZ but also showed a survival benefit in an orthotopic model of glioma. CONCLUSION Andrographolide can enhance anti-tumor activity of TMZ against glioma by inhibiting DKK1 expression.
Collapse
Affiliation(s)
- Zhan-Sheng Zhang
- Department of Pharmacy, The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Zi-Xuan Gao
- Department of Pharmacy, The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Jin-Jin He
- Department of Pharmacy, The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Can Ma
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Hang-Tian Tao
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Feng-Yi Zhu
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Yu-Na Cheng
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Cui-Qing Xie
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Ji-Qin Li
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Zhuang-Zhuang Liu
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Li-Li Hou
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Hua Sun
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China.
| | - Song-Qiang Xie
- Department of Pharmacy, The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China.
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China.
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Kaifeng, 475004, China.
| | - Dong Fang
- Department of Pharmacy, The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China.
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China.
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, Kaifeng, 475004, China.
| |
Collapse
|
2
|
Barakat WEM, Moawed FSM, Ahmed ESA, Abo-Zaid OAR. The hepatotoxicity of γ-radiation synthesized 5-fluorouracil nanogel versus 5-fluorouracil in rats model. Int J Immunopathol Pharmacol 2024; 38:3946320241227099. [PMID: 38207276 PMCID: PMC10785744 DOI: 10.1177/03946320241227099] [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: 09/29/2023] [Accepted: 01/02/2023] [Indexed: 01/13/2024] Open
Abstract
INTRODUCTION The clinical use of 5-fluorouracil (5-FU), a routinely used chemotherapy medication, has a deleterious impact on the liver. Therefore, it is necessary to find a less harmful alternative to minimize liver damage. This study was designed to see how 5-fluorouracil nanogel influenced 5-FU-induced liver damage in rats. METHODS To induce liver damage, male albino rats were injected intraperitoneally with 5-FU (12.5 mg/kg) three doses/week for 1 month. The histopathological examination together with measuring the activities of serum alanine and aspartate aminotransferase enzymes (ALT and AST) were used to evaluate the severity of liver damage besides, hepatic oxidative stress and antioxidant markers were also measured. The hepatic gene expression of heme oxygenase-1 (HO-1), nuclear factor erythroid 2-related factor 2 (Nrf2) and its inhibitor Kelch-like ECH-associated protein-1(Keap-1) in addition to hepatic inflammatory mediators including tumor necrosis factor-α (TNF- α) and interleukins (IL-1β, IL-6) were detected. RESULTS 5-Fu nanogel effectively attenuated 5-FU-induced liver injury by improving the hepatic structure and function (ALT and AST) besides the suppression of the hepatic inflammatory mediators (TNF- α, IL-1β and IL-6). Additionally, 5-FU nanogel alleviated the impaired redox status and restored the antioxidant system via maintaining the cellular homeostasis Keap-1/Nrf2/HO-1 pathway. CONCLUSION Consequently, 5-Fu nanogel exhibited lower liver toxicity compared to 5-FU, likely due to the alleviation of hepatic inflammation and the regulation of the cellular redox pathway.
Collapse
Affiliation(s)
- Wael EM Barakat
- Biochemistry and Molecular Biology Department, Benha University Faculty of Veterinary Medicine, Benha, Egypt
| | - Fatma SM Moawed
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Esraa SA Ahmed
- Radiation Biology Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Omayma AR Abo-Zaid
- Biochemistry and Molecular Biology Department, Benha University Faculty of Veterinary Medicine, Benha, Egypt
| |
Collapse
|
3
|
Yalcın T, Kaya S, Kuloğlu T. Resveratrol may dose-dependently modulate nephrin and OTULIN levels in a doxorubicin-induced nephrotoxicity model. Toxicol Mech Methods 2024; 34:98-108. [PMID: 37807854 DOI: 10.1080/15376516.2023.2268717] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
One of the most important side effects of Doxorubicin (DOX), a chemotherapeutic agent, is nephrotoxicity. The purpose of this study is to determine whether different doses of natural polyphenol Resveratrol (RSV) show antioxidative, anti-inflammatory or antiapoptotic effects in kidney tissue in DOX-induced nephrotoxicity and to detect how nephrin and OTULIN levels are affected in this process. A total of six equal groups made up of the 42 Sprague-Dawley rats utilized in the study (n = 7) were randomly assigned. Except for the control group (no treatment), all treatments were given intraperitoneally to the DOX (15 mg/kg), DOX + RSV I (15 mg/kg DOX+ 1 mg/kg/day RSV), DOX + RSV II (15 mg/kg DOX+ 5 mg/kg/day RSV), RSV I and RSV II groups. Kidney tissues taken from rats sacrificed on the fifteenth day were analyzed biochemically, histologically and immunohistochemically. Accordingly, it was determined that nephrin and OTULIN levels decreased in kidney tissue in DOX-induced nephrotoxicity. Furthermore, DOX caused oxidative stress, inflammation, and apoptosis, as well as histopathological changes in kidney tissue. However, it was observed that DOX-induced changes were regulated by RSV application. RSV was demonstrated to have antioxidant, anti-inflammatory and anti-apoptotic properties in dose-dependent DOX-induced nephrotoxicity. RSV may exert nephroprotective effects by modulating DOX-induced altered nephrin and OTULIN levels.
Collapse
Affiliation(s)
- Tuba Yalcın
- Vocational School of Healthcare Studies, Batman University, Batman, Turkey
| | - Sercan Kaya
- Vocational School of Healthcare Studies, Batman University, Batman, Turkey
| | - Tuncay Kuloğlu
- Department of Histology and Embryology, Faculty of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
4
|
Semenescu AD, Moacă EA, Iftode A, Dehelean CA, Tchiakpe-Antal DS, Vlase L, Vlase AM, Muntean D, Chioibaş R. Phytochemical and Nutraceutical Screening of Ethanol and Ethyl Acetate Phases of Romanian Galium verum Herba ( Rubiaceae). Molecules 2023; 28:7804. [PMID: 38067535 PMCID: PMC10707836 DOI: 10.3390/molecules28237804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Galium species are used worldwide for their antioxidant, antibacterial, antifungal, and antiparasitic properties. Although this plant has demonstrated its antitumor properties on various types of cancer, its biological activity on cutaneous melanoma has not been established so far. Therefore, the present study was designed to investigate the phytochemical profile of two extracts of G. verum L. herba (ethanolic and ethyl acetate) as well as the biological profile (antioxidant, antimicrobial, and antitumor effects) on human skin cancer. The extracts showed similar FT-IR phenolic profiles (high chlorogenic acid, isoquercitrin, quercitrin, and rutin), with high antioxidant capacity (EC50 of ethyl acetate phase (0.074 ± 0.01 mg/mL) > ethanol phase (0.136 ± 0.03 mg/mL)). Both extracts showed antimicrobial activity, especially against Gram-positive Streptococcus pyogenes and Staphylococcus aureus bacilli strains, the ethyl acetate phase being more active. Regarding the in vitro antitumor test, the results revealed a dose-dependent cytotoxic effect against A375 melanoma cell lines, more pronounced in the case of the ethyl acetate phase. In addition, the ethyl acetate phase stimulated the proliferation of human keratinocytes (HaCaT), while this effect was not evident in the case of the ethanolic phase at 24 h post-stimulation. Consequently, G. verum l. could be considered a promising phytocompound for the antitumor approach of cutaneous melanoma.
Collapse
Affiliation(s)
- Alexandra-Denisa Semenescu
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.-D.S.); (E.-A.M.); (C.-A.D.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Elena-Alina Moacă
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.-D.S.); (E.-A.M.); (C.-A.D.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Andrada Iftode
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.-D.S.); (E.-A.M.); (C.-A.D.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Cristina-Adriana Dehelean
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.-D.S.); (E.-A.M.); (C.-A.D.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Diana-Simona Tchiakpe-Antal
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Laurian Vlase
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8th Victor Babes Street, 400347 Cluj-Napoca, Romania;
| | - Ana-Maria Vlase
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8th Victor Babes Street, 400347 Cluj-Napoca, Romania;
| | - Delia Muntean
- Department of Microbiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
- Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Raul Chioibaş
- Department of Surgery I, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timișoara, Romania;
- CBS Medcom Hospital, 12th Popa Sapca Street, 300047 Timisoara, Romania
| |
Collapse
|
5
|
Kapoor S, Gupta N, Sharma A. A Polyherbal Ashwagandha Formulation Exhibits Adjunctive Antitumor Efficacy Against U266 Myeloma Cells by Multi-Strategic Cytotoxic Effects: An Experimental Approach. Asian Pac J Cancer Prev 2023; 24:3705-3714. [PMID: 38019228 PMCID: PMC10772745 DOI: 10.31557/apjcp.2023.24.11.3705] [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: 04/17/2023] [Accepted: 11/04/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND The present study explored the molecular mechanism of herbal (Unani) drug Habb-e-asgandh as anti-tumorigenic adjuvant therapy experimentally in U266 cells and its role in treatment of Multiple myeloma. The formulation of Habb-e-asgandh is investigated alone or as a combinatorial therapy with standard drug lenalidomide to check for its efficacy against U266 myeloma cells for prevention of drug relapse and resistance. METHODS We performed the following assays on singly or in combination of Habb-e-asgandh-Lenalidomide treated U266 cells. The cytotoxicity evaluation done by MTT assay, we studied cell cycle kinetics by Propidium Iodide staining, mitochondrial apoptosis analysis by Annexin V/PI dual staining and JC1 staining assays. Further, anti-oxidative potential was assessed by ORAC assay and cytokine levels estimation of anti-inflammatory (TNF-alpha and IL6) and anti-angiogenic (VEGF and Ang-2) markers were done by ELISA. RESULTS The myeloma U266 cells when treated with Habb-e-asgandh alone or in combination with standard drug lenalidomide showed cytotoxicity in dose dependent manner with promising effects at 0.4 mg/ml (IC30) and 1.5 mg/ml (IC50) inhibitory concentrations. The formulation treated cells showed modulation in cell cycle kinetics patterned by sub Go/G1 population accumulation. Furthermore, it induced mitochondrial apoptosis mainly at half maximal inhibitory concentration and in combinatorial combinations. Significantly elevated oxidative capacities (p<0.05) and reduced levels of angiogenic and pro-inflammatory markers were observed. Multiple mechanism based inhibition by Habb-e-asgandh in co-treatment with lenalidomide against myeloma cells is indicated. Conclusion: Habb-e-asgandh formulation possess anti-tumorigenic efficacy against multiple myeloma. The adjunctive Habb-e-asgandh formulation with standard chemotherapeutic drug may prove to be a potent anti-myeloma agent in interventional therapy for Multiple myeloma if further studied in future avenues.
Collapse
|
6
|
Kavaz D, Faraj REKE. Investigation of composition, antioxidant, antimicrobial and cytotoxic characteristics from Juniperus sabina and Ferula communis extracts. Sci Rep 2023; 13:7193. [PMID: 37137993 PMCID: PMC10156702 DOI: 10.1038/s41598-023-34281-x] [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: 04/13/2022] [Accepted: 04/27/2023] [Indexed: 05/05/2023] Open
Abstract
Plants have been one the most valuable sources of biologically active compounds. This study investigates the chemical composition, as well as the antioxidant, antimicrobial, and cytotoxic activities of methanolic and ethanolic extracts from Juniperus sabina and Ferula communis leaves, grown in Cyprus. Total phenolic and flavonoids content of methanol and ethanol extracts were quantified. Chemical constituents of the leaf extracts were analysed using gas chromatography/mass spectrometry (GC/MS). Mome inositol was the predominant component in the J. Sabina's extracts. The most dominant component in F. communis ethanolic extract was phytol, while in FCL methanolic extract 1,3,4,5 tetrahydroxycyclohexanecarboxylic acid. Antioxidant activities were evaluated by 1, 1-diphenyl-2-picrylhydrazyl (DPPH) free radical-scavenging ability. Antioxidant activity results revealed concentration dependent activity for methanolic and ethanolic extracts from the plant leaves. Antibacterial activity of plant extracts was tested against Gram-negative and Gram-positive bacteria using disk diffusion and minimal inhibitory concentration methods. Cytotoxic activity of plant extracts were evaluated on MCF-7 and MDA-MB-231 breast cancer cell lines, where they demonstrated their potential on the viability of both cell lines. The biological activity revealed by plants is due to the bioactive compounds found in the extracts. These bioactive components could be used as anticancer drug candidates.
Collapse
Affiliation(s)
- Doga Kavaz
- Department of Bioengineering, Cyprus International University, Northern Cyprus Via Mersin 10, Haspolat, Nicosia, Turkey.
- Biotechnology Research Centre, Cyprus International University, Northern Cyprus Via Mersin 10, Haspolat, Nicosia, Turkey.
| | - Razan El Khaled El Faraj
- Department of Bioengineering, Cyprus International University, Northern Cyprus Via Mersin 10, Haspolat, Nicosia, Turkey
| |
Collapse
|
7
|
Exploring the Antiovarian Cancer Mechanisms of Salvia Miltiorrhiza Bunge by Network Pharmacological Analysis and Molecular Docking. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7895246. [PMID: 36483919 PMCID: PMC9726254 DOI: 10.1155/2022/7895246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/13/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022]
Abstract
Background Ovarian cancer was one of the gynecological malignant tumors. Salvia miltiorrhiza Bunge (SMB) was a kind of herbal medicine with an antitumor effect. However, the inhibitory effect of SMB on ovarian cancer and its potential mechanism were still unclear. Objective The antitumor effect of SMB on ovarian cancer was studied by network pharmacology and molecular docking techniques, and its possible molecular mechanisms were analyzed. Method The active ingredients of SMB and the target data of ovarian cancer were obtained from the Traditional Chinese Medicines for Systems Pharmacology Database (TCMSP) and the GeneCards database. The relationship between active ingredients of SMB and ovarian cancer targets was analyzed by String database, David 6.8 online database, and Cytoscape 3.7.2 software, and then potential pathways were screened out. In addition, molecular docking technology was used to verify further the binding effect of antiovarian cancer pathway targets with active ingredients of SMB. Finally, survival analysis was performed for all potential targets. Results We analyzed 71 SMB-ovarian cancer common targets, and the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the PI3K-Akt signaling pathway might be an essential pathway for SMB to inhibit ovarian cancer. Luteolin, Tanshinone IIA, and Cryptotanshinone in SMB might play an important role. HSP90AA1, CDK2, and PIK3CG might be potential targets of SMB in inhibiting ovarian cancer. Conclusion Through network pharmacology and molecular docking analysis, we found that SMB might partially inhibit ovarian cancer by the PI3K-Akt signaling pathway. We believe that SMB might be a potential therapeutic agent for ovarian cancer patients.
Collapse
|
8
|
Cayetano-Salazar L, Nava-Tapia DA, Astudillo-Justo KD, Arizmendi-Izazaga A, Sotelo-Leyva C, Herrera-Martinez M, Villegas-Comonfort S, Navarro-Tito N. Flavonoids as regulators of TIMPs expression in cancer: Consequences, opportunities, and challenges. Life Sci 2022; 308:120932. [PMID: 36067841 DOI: 10.1016/j.lfs.2022.120932] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 12/14/2022]
Abstract
Cancer is one of the leading causes of death in patients worldwide, where invasion and metastasis are directly responsible for this statement. Although cancer therapy has progressed in recent years, current therapeutic approaches are ineffective due to toxicity and chemoresistance. Therefore, it is essential to evaluate other treatment options, and natural products are a promising alternative as they show antitumor properties in different study models. This review describes the regulation of tissue inhibitors of metalloproteinases (TIMPs) expression and the role of flavonoids as molecules with the antitumor activity that targets TIMPs therapeutically. These inhibitors regulate tissue extracellular matrix (ECM) turnover; they inhibit matrix metalloproteinases (MMPs), cell migration, invasion, and angiogenesis and induce apoptosis in tumor cells. Data obtained in cell lines and in vivo models suggest that flavonoids are chemopreventive and cytotoxic against various types of cancer through several mechanisms. Flavonoids also regulate crucial signaling pathways such as focal adhesion kinase (FAK), phosphatidylinositol-3-kinase (PI3K)-Akt, signal transducer and activator of transcription 3 (STAT3), nuclear factor κB (NFκB), and mitogen-activated protein kinase (MAPK) involved in cancer cell migration, invasion, and metastasis. All these data reposition flavonoids as excellent candidates for use in cancer therapy.
Collapse
Affiliation(s)
- Lorena Cayetano-Salazar
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - Dania A Nava-Tapia
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - Kevin D Astudillo-Justo
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - Adán Arizmendi-Izazaga
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - César Sotelo-Leyva
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - Mayra Herrera-Martinez
- Instituto de Farmacobiología, Universidad de la Cañada, Teotitlán de Flores Magón, OAX 68540, Mexico
| | - Sócrates Villegas-Comonfort
- División de Ciencias Naturales e Ingeniería, Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, CDMX 05348, Mexico
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| |
Collapse
|
9
|
Adetunji TL, Olawale F, Olisah C, Adetunji AE, Aremu AO. Capsaicin: A Two-Decade Systematic Review of Global Research Output and Recent Advances Against Human Cancer. Front Oncol 2022; 12:908487. [PMID: 35912207 PMCID: PMC9326111 DOI: 10.3389/fonc.2022.908487] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/30/2022] [Indexed: 12/23/2022] Open
Abstract
Capsaicin (8-methyl-N-vanillyl-6-nonenamide) is one of the most important natural products in the genus Capsicum. Due to its numerous biological effects, there has been extensive and increasing research interest in capsaicin, resulting in increased scientific publications in recent years. Therefore, an in-depth bibliometric analysis of published literature on capsaicin from 2001 to 2021 was performed to assess the global research status, thematic and emerging areas, and potential insights into future research. Furthermore, recent research advances of capsaicin and its combination therapy on human cancer as well as their potential mechanisms of action were described. In the last two decades, research outputs on capsaicin have increased by an estimated 18% per year and were dominated by research articles at 93% of the 3753 assessed literature. In addition, anti-cancer/pharmacokinetics, cytotoxicity, in vivo neurological and pain research studies were the keyword clusters generated and designated as thematic domains for capsaicin research. It was evident that the United States, China, and Japan accounted for about 42% of 3753 publications that met the inclusion criteria. Also, visibly dominant collaboration nodes and networks with most of the other identified countries were established. Assessment of the eligible literature revealed that the potential of capsaicin for mitigating cancer mainly entailed its chemo-preventive effects, which were often linked to its ability to exert multi-biological effects such as anti-mutagenic, antioxidant and anti-inflammatory activities. However, clinical studies were limited, which may be related to some of the inherent challenges associated with capsaicin in the limited clinical trials. This review presents a novel approach to visualizing information about capsaicin research and a comprehensive perspective on the therapeutic significance and applications of capsaicin in the treatment of human cancer.
Collapse
Affiliation(s)
- Tomi Lois Adetunji
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Femi Olawale
- Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban, South Africa
| | - Chijioke Olisah
- Department of Botany and Institute for Coastal and Marine Research, Nelson Mandela University, Port Elizabeth, South Africa
| | | | - Adeyemi Oladapo Aremu
- Indigenous Knowledge Systems Centre, Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho, South Africa
- School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
10
|
Khalaf HM, Hafez SMNA, Abdalla AM, Welson NN, Abdelzaher WY, Abdelbaky FAF. Role of Platelet-activating factor and HO-1 in mediating the protective effect of rupatadine against 5-fluorouracil-induced hepatotoxicity in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:40190-40203. [PMID: 35119631 PMCID: PMC9120097 DOI: 10.1007/s11356-022-18899-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/23/2022] [Indexed: 05/06/2023]
Abstract
5-fluorouracil (5-FU) is a widely used chemotherapeutic drug, but its hepatotoxicity challenges its clinical use. Thus, searching for a hepatoprotective agent is highly required to prevent the accompanied hepatic hazards. The current study aimed to investigate the potential benefit and mechanisms of action of rupatadine (RU), a Platelet-activating factor (PAF) antagonist, in the prevention of 5-FU-related hepatotoxicity in rats. Hepatotoxicity was developed in male albino rats by a single 5-FU (150 mg/kg) intra-peritoneal injection on the 7th day of the experiment. RU (3 mg/kg/day) was orally administrated to the rodents for 10 days. Hepatic toxicity was assessed by measuring both liver and body weights, serum alanine aminotransferase and aspartate aminotransferase (ALT and AST), hepatic oxidative stress parameters (malondialdehyde (MDA), nitric oxide levels (NOx), reduced glutathione (GSH), superoxide dismutase (SOD)), and heme oxygenase-1 (HO-1). Inflammatory markers expressions (inducible nitric oxide synthase (iNOS), tumor necrosis factor-alpha (TNFα), interleukins; IL-1B, IL-6), the apoptotic marker (caspase-3), and PAF were measured in the hepatic tissue. 5-FU-induced hepatotoxicity was proved by the biochemical along with histopathological assessments. RU ameliorated 5-FU-induced liver damage as proved by the improved serum ALT, AST, and hepatic oxidative stress parameters, the attenuated expression of hepatic pro-inflammatory cytokines and PAF, and the up-regulation of HO-1. Therefore, it can be concluded that RU pretreatment exerted a hepatoprotective effect against 5-FU-induced liver damage through both its powerful anti-inflammatory, antioxidant, and anti-apoptotic effect.
Collapse
Affiliation(s)
| | | | | | - Nermeen N. Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | | | | |
Collapse
|
11
|
Markowski A, Jaromin A, Migdał P, Olczak E, Zygmunt A, Zaremba-Czogalla M, Pawlik K, Gubernator J. Design and Development of a New Type of Hybrid PLGA/Lipid Nanoparticle as an Ursolic Acid Delivery System against Pancreatic Ductal Adenocarcinoma Cells. Int J Mol Sci 2022; 23:5536. [PMID: 35628352 PMCID: PMC9143619 DOI: 10.3390/ijms23105536] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/08/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
Despite many attempts, trials, and treatment procedures, pancreatic ductal adenocarcinoma (PDAC) still ranks among the most deadly and treatment-resistant types of cancer. Hence, there is still an urgent need to develop new molecules, drugs, and therapeutic methods against PDAC. Naturally derived compounds, such as pentacyclic terpenoids, have gained attention because of their high cytotoxic activity toward pancreatic cancer cells. Ursolic acid (UA), as an example, possesses a wide anticancer activity spectrum and can potentially be a good candidate for anti-PDAC therapy. However, due to its minimal water solubility, it is necessary to prepare an optimal nano-sized vehicle to overcome the low bioavailability issue. Poly(lactic-co-glycolic acid) (PLGA) polymeric nanocarriers seem to be an essential tool for ursolic acid delivery and can overcome the lack of biological activity observed after being incorporated within liposomes. PLGA modification, with the addition of PEGylated phospholipids forming the lipid shell around the polymeric core, can provide additional beneficial properties to the designed nanocarrier. We prepared UA-loaded hybrid PLGA/lipid nanoparticles using a nanoprecipitation method and subsequently performed an MTT cytotoxicity assay for AsPC-1 and BxPC-3 cells and determined the hemolytic effect on human erythrocytes with transmission electron microscopic (TEM) visualization of the nanoparticles and their cellular uptake. Hybrid UA-loaded lipid nanoparticles were also examined in terms of their stability, coating dynamics, and ursolic acid loading. We established innovative and repeatable preparation procedures for novel hybrid nanoparticles and obtained biologically active nanocarriers for ursolic acid with an IC50 below 20 µM, with an appropriate size for intravenous dosage (around 150 nm), high homogeneity of the sample (below 0.2), satisfactory encapsulation efficiency (up to 70%) and excellent stability. The new type of hybrid UA-PLGA nanoparticles represents a further step in the development of potentially effective PDAC therapies based on novel, biologically active, and promising triterpenoids.
Collapse
Affiliation(s)
- Adam Markowski
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Paweł Migdał
- Polish Academy of Science Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114 Wrocław, Poland; (P.M.); (K.P.)
- Department of Environment Hygiene and Animal Welfare, Bee Division, Wroclaw University of Environmental and Life Sciences, Chelmońskiego 38C, 51-630 Wrocław, Poland
| | - Ewa Olczak
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Adrianna Zygmunt
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Magdalena Zaremba-Czogalla
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| | - Krzysztof Pawlik
- Polish Academy of Science Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114 Wrocław, Poland; (P.M.); (K.P.)
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland; (A.J.); (E.O.); (A.Z.); (M.Z.-C.)
| |
Collapse
|
12
|
Sayed AM, El-Hawary SS, Abdelmohsen UR, Ghareeb MA. Antiproliferative potential of Physalis peruviana-derived magnolin against pancreatic cancer: a comprehensive in vitro and in silico study. Food Funct 2022; 13:11733-11743. [DOI: 10.1039/d2fo01915a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Physalis peruviana L. is a common edible fruit in Egypt and other regional countries.
Collapse
Affiliation(s)
- Ahmed M. Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt
| | - Seham S. El-Hawary
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minia 61111, Egypt
| | - Mosad A. Ghareeb
- Medicinal Chemistry Department, Theodor Bilharz Research Institute, Kornaish El-Nile, Warrak El-Hadar, Imbaba (P.O. 30), Giza 12411, Egypt
| |
Collapse
|
13
|
Ibrahim Fouad G, Ahmed KA. The protective impact of berberine against doxorubicin-induced nephrotoxicity in rats. Tissue Cell 2021; 73:101612. [PMID: 34371291 DOI: 10.1016/j.tice.2021.101612] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 07/29/2021] [Indexed: 01/23/2023]
Abstract
Doxorubicin (DOX) is a well-known anti-neoplastic agent that is widely employed to treat several types of malignancies. The current study was designed to investigate the renoprotective potential of berberine (BEB) on the doxorubicin (DOX)-induced nephrotoxicity and renal fibrosis. Rats were allocated into four groups; Negative Control, DOX nephrotoxic-induced group received a single dose of DOX (20 mg/kg, i.p.), BEB-group received (50 mg/kg, p.o.) for 14 days, and co-treatment group BEB + DOX where rats were pre-treated with BEB for 10 successive days, then received a single dose of DOX on the 11th day, followed by 4 days of receiving BEB. DOX resulted in nephrotoxicity manifested by significant increments in urea, creatinine, and kidney injury molecule (KIM-1), these biochemical findings were supported with the histopathological lesions in renal tissues. Moreover, DOX provoked oxidative stress through enhancing renal malondialdehyde (MDA) and hydrogen peroxide (H2O2) contents, and decreased renal catalase (CAT) activity. DOX triggered renal fibrosis represented by increased transforming growth factor beta-1 (TGF-β1) and elevated collagen deposition. DOX stimulated apoptosis and inflammation in renal tissues as confirmed by increased immunoexpression of caspase-3 and NF-κB, respectively. These effects were alleviated by BEB co-treatment. Co-treatment with BEB markedly prohibited DOX-induced oxidative damage, inflammation, apoptosis, and fibrosis in renal tissue. Histopathological and immunohistochemical investigations showed the nephroprotective potential of BEB on renal injury, which was consistent with the biochemical findings. Accordingly, it could be concluded that the nephroprotective potential of BEB against DOX-induced kidney injury and fibrosis might be mediated by the anti-oxidant, anti-inflammatory and anti-fibrosis activities.
Collapse
Affiliation(s)
- Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt.
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| |
Collapse
|
14
|
Evaluation of the In Vitro Cytotoxic Activity of Ursolic Acid PLGA Nanoparticles against Pancreatic Ductal Adenocarcinoma Cell Lines. MATERIALS 2021; 14:ma14174917. [PMID: 34501007 PMCID: PMC8434451 DOI: 10.3390/ma14174917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022]
Abstract
Among all the types of cancer, Pancreatic Ductal Adenocarcinoma remains one of the deadliest and hardest to fight and there is a critical unmet need for new drugs and therapies for its treatment. Naturally derived compounds, such as pentacyclic triterpenoids, have gathered attention because of their high cytotoxic potential towards pancreatic cancer cells, with a wide biological activity spectrum, with ursolic acid (UA) being one of the most interesting. However, due to its minimal water solubility, it is necessary to prepare a nanocarrier vehicle to aid in the delivery of this compound. Poly(lactic-co-glycolic acid) or PLGA polymeric nanocarriers are an essential tool for ursolic acid delivery and can overcome the lack in its biological activity observed after incorporating within liposomes. We prepared UA-PLGA nanoparticles with a PEG modification, to achieve a long circulation time, by using a nanoprecipitation method and subsequently performed an MTT cytotoxicity assay towards AsPC-1 and BxPC-3 cells, with TEM visualization of the nanoparticles and their cellular uptake. We established repeatable preparation procedures of the nanoparticles and achieved biologically active nanocarriers with an IC50 below 30 µM, with an appropriate size for intravenous dosage (around 140 nm), high sample homogeneity (below 0.2) and reasonable encapsulation efficiency (up to 50%). These results represent the first steps in the development of potentially effective PDAC therapies based on novel biologically active and promising triterpenoids.
Collapse
|
15
|
Ali MS, Lee EB, Lee SJ, Lee SP, Boby N, Suk K, Birhanu BT, Park SC. Aronia melanocarpa Extract Fermented by Lactobacillus plantarum EJ2014 Modulates Immune Response in Mice. Antioxidants (Basel) 2021; 10:antiox10081276. [PMID: 34439524 PMCID: PMC8389331 DOI: 10.3390/antiox10081276] [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: 07/17/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to assess the immunomodulatory effects of fermented Aronia melanocarpa extract (FAME) on RAW 264.7 cells and BALB/c mice. Aronia melanocarpa fruit was fermented with Lactobacillus plantarum EJ2014 by adding yeast extract and monosodium glutamate for 9 days at 30 °C to produce γ-aminobutyric acid (GABA). After fermentation, significant GABA production was noted, along with minerals, polyphenols, and flavonoids (p < 0.05). The polyphenol content was confirmed by liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis. RAW 264.7 cells were stimulated with lipopolysaccharide (LPS, 1 μg/mL) in the presence or absence of FAME, and proinflammatory cytokine contents were measured by qPCR. In the in vivo experiment, female BALB/c mice were administered 125, 250, and 500 mg/kg of FAME for 21 days. FAME treatment increased neutrophil migration and phagocytosis (p < 0.05). It also increased splenocyte proliferation, CD4+ and CD8+ T-cell expression, and lymphocyte proliferation. Furthermore, it increased IFN-γ, IL-2, and IL-4 cytokine levels in a dose-dependent manner (p < 0.05). However, it decreased TNF-α and IL-6 levels (p < 0.05). These results indicate that FAME fortified with GABA including bioactive compounds exerts anti-inflammatory effects by inhibiting proinflammatory cytokines in RAW 264.7 cells and modulates immune response in mice. Thus, FAME could be a potential therapeutic agent for inflammatory disorders.
Collapse
Affiliation(s)
- Md. Sekendar Ali
- Department of Biomedical Science and Department of Pharmacology, School of Medicine, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea; (M.S.A.); (K.S.)
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea; (E.-B.L.); (N.B.)
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong 4318, Bangladesh
| | - Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea; (E.-B.L.); (N.B.)
| | - Seung-Jin Lee
- Development and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Korea;
| | - Sam-Pin Lee
- Department of Food Science and Technology, Keimyung University, Daegu 42601, Korea;
| | - Naila Boby
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea; (E.-B.L.); (N.B.)
| | - Kyoungho Suk
- Department of Biomedical Science and Department of Pharmacology, School of Medicine, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea; (M.S.A.); (K.S.)
| | - Biruk Tesfaye Birhanu
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea; (E.-B.L.); (N.B.)
- Correspondence: (B.T.B.); (S.-C.P.); Tel.: +82-10-5105-5545 (B.T.B.); +82-53-950-5964 (S.-C.P.)
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea; (E.-B.L.); (N.B.)
- Correspondence: (B.T.B.); (S.-C.P.); Tel.: +82-10-5105-5545 (B.T.B.); +82-53-950-5964 (S.-C.P.)
| |
Collapse
|
16
|
Bhargava P, Mahanta D, Kaul A, Ishida Y, Terao K, Wadhwa R, Kaul SC. Experimental Evidence for Therapeutic Potentials of Propolis. Nutrients 2021; 13:2528. [PMID: 34444688 PMCID: PMC8397973 DOI: 10.3390/nu13082528] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/07/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Propolis is produced by honeybees from materials collected from plants they visit. It is a resinous material having mixtures of wax and bee enzymes. Propolis is also known as bee glue and used by bees as a building material in their hives, for blocking holes and cracks, repairing the combs and strengthening their thin borders. It has been extensively used since ancient times for different purposes in traditional human healthcare practices. The quality and composition of propolis depend on its geographic location, climatic zone and local flora. The New Zealand and Brazilian green propolis are the two main kinds that have been extensively studied in recent years. Their bioactive components have been found to possess a variety of therapeutic potentials. It was found that Brazilian green propolis improves the cognitive functions of mild cognitive impairments in patients living at high altitude and protects them from neurodegenerative damage through its antioxidant properties. It possesses artepillin C (ARC) as the key component, also known to possess anticancer potential. The New Zealand propolis contains caffeic acid phenethyl ester (CAPE) as the main bioactive with multiple therapeutic potentials. Our lab performed in vitro and in vivo assays on the extracts prepared from New Zealand and Brazilian propolis and their active ingredients. We provided experimental evidence that these extracts possess anticancer, antistress and hypoxia-modulating activities. Furthermore, their conjugation with γCD proved to be more effective. In the present review, we portray the experimental evidence showing that propolis has the potential to be a candidate drug for different ailments and improve the quality of life.
Collapse
Affiliation(s)
- Priyanshu Bhargava
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (P.B.); (D.M.); (A.K.); (R.W.)
| | - Debajit Mahanta
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (P.B.); (D.M.); (A.K.); (R.W.)
- DBT-APSCS&T Centre of Excellence for Bioresources and Sustainable Development, Kimin 791121, India
| | - Ashish Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (P.B.); (D.M.); (A.K.); (R.W.)
| | - Yoshiyuki Ishida
- CycloChem Co., Ltd., 7-4-5 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan; (Y.I.); (K.T.)
| | - Keiji Terao
- CycloChem Co., Ltd., 7-4-5 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan; (Y.I.); (K.T.)
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (P.B.); (D.M.); (A.K.); (R.W.)
| | - Sunil C. Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba 305-8565, Japan; (P.B.); (D.M.); (A.K.); (R.W.)
- Kaul-Tech Co., Ltd., Nagakunidai 3-24, Tsuchiura 300-0810, Japan
| |
Collapse
|
17
|
Sipos S, Moacă EA, Pavel IZ, Avram Ş, Crețu OM, Coricovac D, Racoviceanu RM, Ghiulai R, Pană RD, Şoica CM, Borcan F, Dehelean CA, Crăiniceanu Z. Melissa officinalis L. Aqueous Extract Exerts Antioxidant and Antiangiogenic Effects and Improves Physiological Skin Parameters. Molecules 2021; 26:molecules26082369. [PMID: 33921730 PMCID: PMC8073307 DOI: 10.3390/molecules26082369] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 11/16/2022] Open
Abstract
Melissa officinalis (MO) is a medicinal plant well-known for its multiple pharmacological effects, including anti-inflammatory, anticancer and beneficial effects on skin recovery. In this context, the present study was aimed to investigate the in vitro and in vivo safety profile of an MO aqueous extract by assessing cell viability on normal (HaCaT-human keratinocytes) and tumor (A375-human melanoma) cells and its impact on physiological skin parameters by a non-invasive method. In addition, the antioxidant activity and the antiangiogenic potential of the extract were verified. A selective cytotoxic effect was noted in A375 cells, while no toxicity was noticed in healthy cells. The MO aqueous extract safety profile after topical application was investigated on SKH-1 mice, and an enhanced skin hydration and decreased erythema and transepidermal water loss levels were observed. The in ovo CAM assay, performed to investigate the potential modulating effect on the angiogenesis process and the blood vessels impact, indicated that at concentrations of 100 and 500 µg/mL, MO aqueous extract induced a reduction of thin capillaries. No signs of vascular toxicity were recorded at concentrations as high as 1000 μg/mL. The aqueous extract of MO leaves can be considered a promising candidate for skin disorders with impaired physiological skin parameters.
Collapse
Affiliation(s)
- Simona Sipos
- Department of Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania;
| | - Elena-Alina Moacă
- Department of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (E.-A.M.); (D.C.); (C.A.D.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (R.-M.R.); (R.G.); (C.M.Ş.)
| | - Ioana Zinuca Pavel
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (R.-M.R.); (R.G.); (C.M.Ş.)
- Department of Pharmacognosy, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania
- Correspondence: (I.Z.P.); (Ş.A.)
| | - Ştefana Avram
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (R.-M.R.); (R.G.); (C.M.Ş.)
- Department of Pharmacognosy, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania
- Correspondence: (I.Z.P.); (Ş.A.)
| | - Octavian Marius Crețu
- Department of Surgery, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania;
| | - Dorina Coricovac
- Department of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (E.-A.M.); (D.C.); (C.A.D.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (R.-M.R.); (R.G.); (C.M.Ş.)
| | - Roxana-Marcela Racoviceanu
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (R.-M.R.); (R.G.); (C.M.Ş.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania
| | - Roxana Ghiulai
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (R.-M.R.); (R.G.); (C.M.Ş.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania
| | - Ramona Daniela Pană
- Department VIII—Neuroscience, Discipline of Medical Deontology. Bioethics, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timisoara, Romania;
| | - Codruţa Marinela Şoica
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (R.-M.R.); (R.G.); (C.M.Ş.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania
| | - Florin Borcan
- Department of Analytical Chemistry, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania;
| | - Cristina Adriana Dehelean
- Department of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (E.-A.M.); (D.C.); (C.A.D.)
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania; (R.-M.R.); (R.G.); (C.M.Ş.)
| | - Zorin Crăiniceanu
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania;
| |
Collapse
|
18
|
Abildgaard C, Rizza S, Christiansen H, Schmidt S, Dahl C, Abdul-Al A, Christensen A, Filomeni G, Guldberg P. Screening of metabolic modulators identifies new strategies to target metabolic reprogramming in melanoma. Sci Rep 2021; 11:4390. [PMID: 33623106 PMCID: PMC7902673 DOI: 10.1038/s41598-021-83796-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
The prognosis of metastatic melanoma remains poor due to de novo or acquired resistance to immune and targeted therapies. Previous studies have shown that melanoma cells have perturbed metabolism and that cellular metabolic pathways represent potential therapeutic targets. To support the discovery of new drug candidates for melanoma, we examined 180 metabolic modulators, including phytochemicals and anti-diabetic compounds, for their growth-inhibitory activities against melanoma cells, alone and in combination with the BRAF inhibitor vemurafenib. Two positive hits from this screen, 4-methylumbelliferone (4-MU) and ursolic acid (UA), were subjected to validation and further characterization. Metabolic analysis showed that 4-MU affected cellular metabolism through inhibition of glycolysis and enhanced the effect of vemurafenib to reduce the growth of melanoma cells. In contrast, UA reduced mitochondrial respiration, accompanied by an increase in the glycolytic rate. This metabolic switch potentiated the growth-inhibitory effect of the pyruvate dehydrogenase kinase inhibitor dichloroacetate. Both drug combinations led to increased production of reactive oxygen species, suggesting the involvement of oxidative stress in the cellular response. These results support the potential use of metabolic modulators for combination therapies in cancer and may encourage preclinical validation and clinical testing of such treatment strategies in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Cecilie Abildgaard
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
- Department of Clinical Genetics, Lillebaelt Hospital - University Hospital of Southern Denmark, Vejle, Denmark
| | - Salvatore Rizza
- Redox Biology Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Helle Christiansen
- Lundbeckfonden Center of Excellence NanoCAN, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Molecular Oncology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Steffen Schmidt
- Lundbeckfonden Center of Excellence NanoCAN, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Molecular Oncology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Christina Dahl
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Ahmad Abdul-Al
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Annette Christensen
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Giuseppe Filomeni
- Redox Biology Group, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Biology, Tor Vergata University of Rome, Rome, Italy
- Center for Healthy Aging, Copenhagen University, Copenhagen, Denmark
| | - Per Guldberg
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark.
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
19
|
Ranneh Y, Akim AM, Hamid HA, Khazaai H, Fadel A, Zakaria ZA, Albujja M, Bakar MFA. Honey and its nutritional and anti-inflammatory value. BMC Complement Med Ther 2021; 21:30. [PMID: 33441127 PMCID: PMC7807510 DOI: 10.1186/s12906-020-03170-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/29/2020] [Indexed: 02/08/2023] Open
Abstract
Inflammation is the main key role in developing chronic diseases including cancer, cardiovascular diseases, diabetes, arthritis, and neurodegenerative diseases which possess a huge challenge for treatment. With massively compelling evidence of the role played by nutritional modulation in preventing inflammation-related diseases, there is a growing interest into the search for natural functional foods with therapeutic and preventive actions. Honey, a nutritional healthy product, is produced mainly by two types of bees: honeybee and stingless bee. Since both types of honey possess distinctive phenolic and flavonoid compounds, there is recently an intensive interest in their biological and clinical actions against inflammation-mediated chronic diseases. This review shed the light specifically on the bioavailability and bioaccessibility of honey polyphenols and highlight their roles in targeting inflammatory pathways in gastrointestinal tract disorders, edema, cancer, metabolic and cardiovascular diseases and gut microbiota.
Collapse
Affiliation(s)
- Yazan Ranneh
- Department of Technology and Natural Resources, Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia, 86400, Pagoh, Johor, Malaysia
| | - Abdah Md Akim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| | - Hasiah Ab Hamid
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Huzwah Khazaai
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Abdulmannan Fadel
- Sport and Exercises Sciences School, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Zainul Amiruddin Zakaria
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Mohammed Albujja
- Department of Forensic Biology, Faculty of Forensic Sciences, Naif Arab University of Security Sciences, Riyadh, 14812, Saudi Arabia
| | - Mohd Fadzelly Abu Bakar
- Department of Technology and Natural Resources, Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia, 86400, Pagoh, Johor, Malaysia
| |
Collapse
|
20
|
Agarwal S, Maekawa T. Nano delivery of natural substances as prospective autophagy modulators in glioblastoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102270. [PMID: 32702467 DOI: 10.1016/j.nano.2020.102270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
Abstract
Glioblastoma is the most destructive type of malignant brain tumor in humans due to cancer relapse. Latest studies have indicated that cancer cells are more reliant on autophagy for survival than non-cancer cells. Autophagy is entitled as programmed cell death type II and studies imply that it is a comeback of cancer cells to innumerable anti-cancer therapies. To diminish the adverse consequences of chemotherapeutics, numerous herbs of natural origin have been retained in cancer treatments. Additionally, autophagy induction occurs via their tumor suppressive actions that could cause cell senescence and increase apoptosis-independent cell death. However, most of the drugs have poor solubility and thus nano drug delivery systems possess excessive potential to improve the aqueous solubility and bioavailability of encapsulated drugs. There is a pronounced need for more therapies for glioblastoma treatment and hereby, the fundamental mechanisms of natural autophagy modulators in glioblastoma are prudently reviewed in this article.
Collapse
Affiliation(s)
- Srishti Agarwal
- Bio-Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan.
| | - Toru Maekawa
- Bio-Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama, Japan
| |
Collapse
|
21
|
Saralamma VVG, Vetrivel P, Lee HJ, Kim SM, Ha SE, Murugesan R, Kim EH, Heo JD, Kim GS. Comparative proteomic analysis uncovers potential biomarkers involved in the anticancer effect of Scutellarein in human gastric cancer cells. Oncol Rep 2020; 44:939-958. [PMID: 32705238 PMCID: PMC7388386 DOI: 10.3892/or.2020.7677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Scutellarein (SCU), a flavone that belongs to the flavonoid family and abundantly present in Scutellaria baicalensis a flowering plant in the family Lamiaceae, has been reported to exhibit anticancer effects in several cancer cell lines including gastric cancer (GC). Although our previous study documented the mechanisms of Scutellarein‑induced cytotoxic effects, the literature shows that the proteomic changes that are associated with the cellular response to SCU have been poorly understood. To avoid adverse side‑effects and significant toxicity of chemotherapy in patients who react poorly, biomarkers anticipating therapeutic responses are imperative. In the present study, we utilized a comparative proteomic analysis to identify proteins associated with Scutellarein (SCU)‑induced cell death in GC cells (AGS and SNU484), by integrating two‑dimensional gel electrophoresis (2‑DE), mass spectrometry (MS), and bioinformatics to analyze the proteins. Proteomic analysis between SCU‑treated and DMSO (control) samples successfully identified 41 (AGS) and 31 (SNU484) proteins by MALDI‑TOF/MS analysis and protein database search. Comparative proteomics analysis between AGS and SNU484 cells treated with SCU revealed a total of 7 protein identities commonly expressed and western blot analysis validated a subset of identified critical proteins, which were consistent with those of the 2‑DE outcome. Molecular docking studies also confirmed the binding affinity of SCU towards these critical proteins. Phosphatidylinositol 4,5‑bisphosphate 3‑kinase catalytic subunit β isoform (PIK3CB) protein expression was accompanied by a distinct group of cellular functions, including cell growth, and proliferation. Cancerous inhibitor of protein phosphatase 2A (CIP2A), is one of the oncogenic molecules that have been shown to promote tumor growth and resistance to apoptosis and senescence‑inducing therapies. In the present study, both PIK3CB and CIP2A proteins were downregulated in SCU‑treated cells, which boosts our previous results of SCU to induce apoptosis and inhibits GC cell growth by regulating these critical proteins. The comparative proteomic analysis has yielded candidate biomarkers of response to SCU treatment in GC cell models and further validation of these biomarkers will help the future clinical development of SCU as a novel therapeutic drug.
Collapse
Affiliation(s)
- Venu Venkatarame Gowda Saralamma
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Preethi Vetrivel
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Ho Jeong Lee
- Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, Jinju, Gyeongnam 52834, Republic of Korea
| | - Seong Min Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Sang Eun Ha
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| | - Rajeswari Murugesan
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641043, India
| | - Eun Hee Kim
- Department of Nursing Science, International University of Korea, Jinju, Gyeongnam 52833, Republic of Korea
| | - Jeong Doo Heo
- Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, Jinju, Gyeongnam 52834, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 52828, Republic of Korea
| |
Collapse
|
22
|
Prša P, Karademir B, Biçim G, Mahmoud H, Dahan I, Yalçın AS, Mahajna J, Milisav I. The potential use of natural products to negate hepatic, renal and neuronal toxicity induced by cancer therapeutics. Biochem Pharmacol 2020; 173:113551. [PMID: 31185225 DOI: 10.1016/j.bcp.2019.06.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022]
|
23
|
Abbas MN, Kausar S, Cui H. Therapeutic potential of natural products in glioblastoma treatment: targeting key glioblastoma signaling pathways and epigenetic alterations. Clin Transl Oncol 2019; 22:963-977. [DOI: 10.1007/s12094-019-02227-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022]
|
24
|
Docetaxel Combined with Thymoquinone Induces Apoptosis in Prostate Cancer Cells via Inhibition of the PI3K/AKT Signaling Pathway. Cancers (Basel) 2019; 11:cancers11091390. [PMID: 31540423 PMCID: PMC6770702 DOI: 10.3390/cancers11091390] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023] Open
Abstract
Toxicity and the development of resistance by cancer cells are impediments for docetaxel (DTX), a primary drug for treating prostate cancer (PCa). Since the combination of DTX with natural compounds can increase its effectiveness by reducing its toxic concentrations, we evaluated a combination of thymoquinone (TQ) with DTX and determined its cytotoxicity against PCa cells (DU145 and C4-2B). This combination, in a concentration-dependent manner, resulted in synergistic cytotoxicity and apoptosis in comparison to either DTX or TQ alone. In addition, inhibition of cell survival pathways by PI3K/AKT inhibitors conferred sensitivity of DU145 and C4-2B cells to the combination as compared to the individual drugs. Moreover, the combined drugs (DTX+TQ) with inhibitors of PI3K/AKT increased the expression of pro-apoptotic markers (BAX and BID) along with caspase-3, PARP and decreased expression of the anti-apoptotic marker, BCL-XL. These data show that, for PCa cells, the cytotoxic effect of the DTX and TQ combination correlates with a block of the PI3K/AKT signaling pathway. These findings indicate that the combination of DTX and TQ, by blocking of the PI3K/AKT pathway, will improve the survival rate and quality of life of PCa patients.
Collapse
|
25
|
Rady H, Salem S, Ez El-Arab M. Primmorph extracts and mesohyls of marine sponges inhibit proliferation and migration of hepatocellular carcinoma cells in vitro. J Pharm Anal 2019; 9:284-291. [PMID: 31452967 PMCID: PMC6704043 DOI: 10.1016/j.jpha.2019.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 01/01/2023] Open
Abstract
Cancer recurrence and severe side effects of currently being used chemotherapeutic agents reduce their clinical efficacy. Thus, there is a constant need to develop alternative anticancer drugs. Sustainable supply is an important challenge facing marine-based drug discovery. Primmorph, a 3D cell culture system, could provide a sustainable source to produce metabolites for anticancer drugs from marine sponges. In the present work, the anticancer activity of primmorph extracts and mesohyls of Negombata magnifica, Hemimycle arabica, Crella spinulata, and Stylissa carteri sponges was evaluated. Antiproliferative activity was studied in terms of cytotoxicity, colony formation, cell cycle, and apoptosis. Migration was assessed by migration assay and matrix metalloproteinase activity. The expression of proliferation and migration-related genes was analyzed using real time PCR. Migration and proliferation activities of HepG2 cells were inhibited by treatment with primmorph extracts and mesohyls of N. magnifica, H. arabica, and C. spinulata. The mesohyl of S. carteri did not show any anticancer activity although the primmorph extract led to cell cycle arrest. Among the selected sponge species, the primmorph extract of C. spinulata was the most promising anticancer agent regarding antiproliferative and antimigratory activities. In addition, primmorph extracts have the advantage of working under well-defined and controlled conditions, which allows the easy application as a bioreactor.
Collapse
Affiliation(s)
- Hanaa Rady
- Chemistry of Natural Compound Department, National Research Centre, Cairo, Egypt
| | - Sohair Salem
- Molecular Genetics and Enzymology Department, National Research Centre, Cairo, Egypt
| | - Mohamed Ez El-Arab
- National Institute of Oceanography and Fisheries (NIOF), Hurghada, Egypt
| |
Collapse
|
26
|
Alsayari A, Muhsinah AB, Hassan MZ, Ahsan MJ, Alshehri JA, Begum N. Aurone: A biologically attractive scaffold as anticancer agent. Eur J Med Chem 2019; 166:417-431. [PMID: 30739824 DOI: 10.1016/j.ejmech.2019.01.078] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022]
Abstract
Aurones are very simple, promising anticancer lead molecules containing three rings (A, B and C). A very slight structural variation in the aurones elicits diverse affinity and specificity towards different molecular targets. The present review discusses the design, discovery and development of natural and synthetic aurones as small molecule anticancer agents. Detailed structure-activity relationship and intermolecular interactions at different targets are also discussed. Due to their rare occurrence in nature and minimal mention in literature, the anticancer potential of aurones is rather recent but in constant progress.
Collapse
Affiliation(s)
| | | | | | | | | | - Naseem Begum
- College of Applied Medical Sciences, King Khalid University, Abha, 62529, Saudi Arabia
| |
Collapse
|
27
|
Zhao X, Wen F, Wang W, Lu Z, Guo Q. Actinidia arguta (Hardy Kiwi) Root Extract Exerts Anti-cancer Effects via Mcl-1-Mediated Apoptosis in Cholangiocarcinoma. Nutr Cancer 2019; 71:246-256. [PMID: 30633583 DOI: 10.1080/01635581.2018.1557218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive and chemoresistant liver malignancy. Thus, identification of strategies to overcome insensitivity to apoptosis and growth inhibition is a growing focus of research in this malignancy. This study evaluated the potential anti-cancer effects of an ethanol extract from the Actinidia arguta (Hardy Kiwi) root (RAE) on CCA. Our data demonstrated that RAE decreased cell viability and induced apoptosis by activation of Caspase 3, Caspase 8, and Poly (ADP-ribose) polymerase (PARP) in two CCA cell lines. RAE induced a decrease in Mcl-1 in cultured CCA cells and in xenograft CCA tumors. Administration of RAE every other day led to significant growth inhibition in tumor burden xenograft CCA mice. Western blotting analysis of paired human CCA and normal adjacent tissues from the same patient revealed that CCA tissues exhibited significantly higher Mcl-1 expression than normal tissues. Taken together, our findings demonstrated the anti-cancer effects of RAE on CCA both in vitro and in vivo. These data suggest that RAE may be a promising anti-CCA agent and could be beneficial in the treatment of CCA through the targeting of Mcl-1.
Collapse
Affiliation(s)
- Xiangxuan Zhao
- a Department of Radiology , Shengjing Hospital of China Medical University , Shenyang , China
| | - Feng Wen
- a Department of Radiology , Shengjing Hospital of China Medical University , Shenyang , China
| | - Wei Wang
- a Department of Radiology , Shengjing Hospital of China Medical University , Shenyang , China
| | - Zaiming Lu
- a Department of Radiology , Shengjing Hospital of China Medical University , Shenyang , China
| | - Qiyong Guo
- a Department of Radiology , Shengjing Hospital of China Medical University , Shenyang , China
| |
Collapse
|
28
|
Kundur S, Prayag A, Selvakumar P, Nguyen H, McKee L, Cruz C, Srinivasan A, Shoyele S, Lakshmikuttyamma A. Synergistic anticancer action of quercetin and curcumin against triple‐negative breast cancer cell lines. J Cell Physiol 2018; 234:11103-11118. [DOI: 10.1002/jcp.27761] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Sai Kundur
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Amrita Prayag
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Priyanga Selvakumar
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Hung Nguyen
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Lloyd McKee
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Clairissa Cruz
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Asha Srinivasan
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Sunday Shoyele
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Ashakumary Lakshmikuttyamma
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| |
Collapse
|
29
|
Agarwal S, Muniyandi P, Maekawa T, Kumar DS. Vesicular systems employing natural substances as promising drug candidates for MMP inhibition in glioblastoma: A nanotechnological approach. Int J Pharm 2018; 551:339-361. [PMID: 30236647 DOI: 10.1016/j.ijpharm.2018.09.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme (GBM), one of the most lethal Brain tumors, characterized by its high invasive nature and increased mortality rates forms a major bottleneck in transport of therapeutics across the Blood Brain Barrier (BBB). Matrix metalloproteinases (MMPs) are classified as enzymes, which are found to be up regulated in the Glioma tumor microenvironment and thus can be considered as a target for inhibition for curbing GBM. Many chemotherapeutics and techniques have been employed for inhibiting MMPs till now but all of them failed miserably and were withdrawn in clinical trials due to their inability in restricting the tumor growth or increasing the overall survival rates. Thus, the quest for finding the suitable MMP inhibitor is still on and there is a critical need for identification of novel compounds which can alter the BBB permeability, restrain tumor growth and prevent tumor recurrence. Currently, naturally derived substances are gaining widespread attention as tumor inhibitors and many studies have been reported by far highlighting their importance in restricting MMP expression thus serving as chemotherapeutics for cancer due to their minimal toxicity. These substances may serve as probable candidates for inhibiting MMP expression in GBM. However, targeting and delivering the inhibitor to its target site is an issue that needs to be overcome in order to attain maximum specificity and sustained release. The birth of nanotechnology served as a boon in delivering drugs to the most complicated areas thus paving way for Nano drug delivery. An efficient Nano carrier with ability to cross the BBB and competently kill the Glioma cells forms the prerequisite for GBM chemotherapy. Vesicular drug delivery systems are one such class of carriers, which have the capacity to release the drug at a predetermined rate at the target site thus minimizing any undesirable side effects. Exploiting vesicular systems as promising Nano drug carriers to formulate naturally derived substances, that can bypass the BBB and act as an inhibitor against MMPs in GBM is the main theme of this review.
Collapse
Affiliation(s)
- Srishti Agarwal
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - Priyadharshni Muniyandi
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - Toru Maekawa
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - D Sakthi Kumar
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan.
| |
Collapse
|
30
|
Ham J, Kim J, Bazer FW, Lim W, Song G. Silibinin‐induced endoplasmic reticulum stress and mitochondrial dysfunction suppress growth of endometriotic lesions. J Cell Physiol 2018; 234:4327-4341. [DOI: 10.1002/jcp.27212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/17/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Jiyeon Ham
- Department of Biotechnology Institute of Animal Molecular Biotechnology Korea University Seoul Republic of Korea
| | - Jonggun Kim
- Department of Biotechnology Institute of Animal Molecular Biotechnology Korea University Seoul Republic of Korea
| | - Fuller W. Bazer
- Department of Animal Science Center for Animal Biotechnology and Genomics, Texas A&M University College Station Texas
| | - Whasun Lim
- Department of Biomedical Sciences Catholic Kwandong University Gangneung Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology Institute of Animal Molecular Biotechnology Korea University Seoul Republic of Korea
| |
Collapse
|
31
|
Piperine functions as a tumor suppressor for human ovarian tumor growth via activation of JNK/p38 MAPK-mediated intrinsic apoptotic pathway. Biosci Rep 2018; 38:BSR20180503. [PMID: 29717031 PMCID: PMC6435525 DOI: 10.1042/bsr20180503] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/23/2018] [Accepted: 04/30/2018] [Indexed: 12/24/2022] Open
Abstract
Piperine, a kind of natural alkaloid found in the fruit of black (Piper nigrum Linn) and long (Piper longum Linn), has shown antitumor activities toward various cancer cell lines. However, the antitumor effects of Piperine on ovarian cancer and the underlying mechanism are not fully elucidated. Our result showed that Piperine reduced the cell viability of A2780 cells in a concentration and time-dependent manner, but has not any effect on normal ovarian cells. Flow cytometric analysis revealed that Piperine suppressed cells proliferation via induction of apoptosis, which was followed by release of mitochondrial cytochrome c to cytosol, activation of caspase-3 and -9, as well as cleaved PARP. Moreover, Western blot results confirmed that Piperine (8, 16, and 20 μM) decreased phosphorylation of JNK and p38 MAPK in A2780 cells. In addition, caspase-3 inhibitor (Z-DEVD-FMK), caspase-9 inhibitor (Z-LEDH-FMK), JNK-inhibitor (SP600125), or p38 MAPK inhibitor (SB203580) could abate the apoptosis induced by Piperine (20 μM) treatment, while caspase-8 inhibitor (Z-IETD- FMK) exhibited no inhibitory effect on the induction of apoptosis in A2780 cells. These results provide the first evidence for the anticancer potential of Piperine in ovarian cancer cells, partially via JNK/p38 MAPK-mediated intrinsic apoptotic pathway.
Collapse
|
32
|
Lukša J, Vepštaitė-Monstavičė I, Yurchenko V, Serva S, Servienė E. High content analysis of sea buckthorn, black chokeberry, red and white currants microbiota - A pilot study. Food Res Int 2018; 111:597-606. [PMID: 30007724 DOI: 10.1016/j.foodres.2018.05.060] [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: 02/23/2018] [Revised: 05/04/2018] [Accepted: 05/23/2018] [Indexed: 01/22/2023]
Abstract
The high potential of sea buckthorn, black chokeberry, red and white currants in healthy food industry boosted interest in the plant cultivation. The present study is the first work providing comprehensive information on microbial populations of these berries. Next Generation Sequencing allowed identification of eukaryotic and prokaryotic microorganisms prevalent on specific berries, including uncultivable microorganisms. Our study revealed the broad diversity of berries-associated bacterial and fungal microorganisms. Analysis of representative microbial OTUs showed a clear separation among inhabitants of sea buckthorn, black chokeberry and both currants, indicating plant-defined differences in the composition of the bacterial and fungal microbiota. Among the microorganisms distributed on tested berries, we documented potentially beneficial fungi and bacteria along with potential phytopathogens or those harmful for humans. Thus, plant microbiota appears to be highly relevant for the evaluation of the microbiota impact on food quality and human health.
Collapse
Affiliation(s)
- Juliana Lukša
- Laboratory of Genetics, Institute of Botany, Nature Research Centre, Akademijos str. 2, Vilnius LT-08412, Lithuania
| | - Iglė Vepštaitė-Monstavičė
- Laboratory of Genetics, Institute of Botany, Nature Research Centre, Akademijos str. 2, Vilnius LT-08412, Lithuania
| | - Vyacheslav Yurchenko
- Life Science Research Centre and Institute of Environmental Technologies, Faculty of Science, University of Ostrava, Chittussiho 10, 710 00 Ostrava, Czech Republic
| | - Saulius Serva
- Department of Biochemistry and Molecular Biology, Institute of Biosciences, Vilnius University, Saulėtekio al.7, Vilnius LT-10257, Lithuania
| | - Elena Servienė
- Laboratory of Genetics, Institute of Botany, Nature Research Centre, Akademijos str. 2, Vilnius LT-08412, Lithuania.
| |
Collapse
|
33
|
Doan P, Anufrieva O, Yli-Harja O, Kandhavelu M. In vitro characterization of alkylaminophenols-induced cell death. Eur J Pharmacol 2017; 820:229-234. [PMID: 29275157 DOI: 10.1016/j.ejphar.2017.12.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/30/2022]
Abstract
Alkylaminophenols are synthetic derivatives well known for their anticancer activity. In the previous studies, we described the activity of the series of Alkylaminophenols derivatives and their ability to induce cell death for many cancer cell lines. However, temporal heterogeneity in cell death induced by lead compounds, N-(2-hydroxy-5-nitrophenyl (4'-methylphenyl) methyl) indoline (Compound I) and 2-((3,4-dihydroquinolin-1(2H)-yl) (4-methoxyphenyl) methyl) phenol (Compound II), has never been tested on osteosarcoma cells (U2OS). Here, we address the level of cell-to-cell heterogeneity by examine whether differences in the type of compounds could influence its effects on cell death of U2OS. Here, we applied imaging, computational methods and biochemical methods to study heterogeneity, apoptosis, reactive oxygen species and caspase. Our results demonstrate that the Hill coefficient of dose-response curve of Compound II is greater than compound I in treated U2OS cells. Both Compounds trigger not only apoptotic cell death but also necro-apoptotic and necrotic cell death. The percentage of these sub-populations varies depending on compounds in which greater variance is induced by compound II than Compound I. We also identified the accumulation of compounds-induced reactive oxygen species during the treatment. This resulted in caspase 3/7 activation in turn induced apoptosis. In summary, the screening of Compound I and II molecules for heterogeneity, apoptosis, reactive oxygen species and caspase has identified compound II as promising anti-osteosarcoma cancer agent. Compound II could be a promising lead compound for future antitumor agent development.
Collapse
Affiliation(s)
- Phuong Doan
- Molecular Signaling Lab, Computational Systems Biology Research Group, BioMediTech and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, P.O.Box 553, 33101 Tampere, Finland
| | - Olga Anufrieva
- Molecular Signaling Lab, Computational Systems Biology Research Group, BioMediTech and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, P.O.Box 553, 33101 Tampere, Finland
| | - Olli Yli-Harja
- Molecular Signaling Lab, Computational Systems Biology Research Group, BioMediTech and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, P.O.Box 553, 33101 Tampere, Finland; Institute for Systems Biology, 1441N 34th Street, Seattle, WA 98103-8904, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Computational Systems Biology Research Group, BioMediTech and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, P.O.Box 553, 33101 Tampere, Finland.
| |
Collapse
|
34
|
Liu W, Zhang B, Chen G, Wu W, Zhou L, Shi Y, Zeng Q, Li Y, Sun Y, Deng X, Wang F. Targeting miR-21 with Sophocarpine Inhibits Tumor Progression and Reverses Epithelial-Mesenchymal Transition in Head and Neck Cancer. Mol Ther 2017; 25:2129-2139. [PMID: 28571917 DOI: 10.1016/j.ymthe.2017.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 12/20/2022] Open
Abstract
A major challenge for cancer chemotherapy is the development of safe and clinically effective chemotherapeutic agents. With its low toxicity profile, sophocarpine (SC), a naturally occurring tetracyclic quinolizidine alkaloid derived from Sophora alopecuroides L, has shown promising therapeutic properties, including anti-inflammatory, anti-nociceptive, and antivirus activities. However, the antitumor efficacy of SC and its underlying mechanisms have not been completely delineated. In the present study, the inhibitory effect of SC on head and neck squamous cell carcinoma (HNSCC) progression and possible mechanisms for this effect involving microRNA-21 (miR-21) regulation were investigated. By cell viability, Transwell, and wound healing assays, we show that SC effectively inhibited proliferation, invasion, and migration of HNSCC cells. Moreover, SC exerted its growth-inhibitory effect via the downregulation of miR-21 expression by blocking Dicer-mediated miR-21 maturation. Furthermore, SC treatment led to the increased expression of PTEN and p38MAPK phosphorylation as well as the reversal of epithelial-mesenchymal transition (EMT), which was rescued by ectopic expression of miR-21 in cells. Notably, SC dramatically repressed tumor growth without observable tissue cytotoxicity in a mouse xenograft model of HNSCC. Our findings offer a preclinical proof of concept for SC as a leading natural agent for HNSCC cancer therapy.
Collapse
Affiliation(s)
- Wei Liu
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China; Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Beilei Zhang
- Department of Gynecology and Obstetrics, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Guo Chen
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Wenjiao Wu
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
| | - Lin Zhou
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
| | - Yaru Shi
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
| | - Qi Zeng
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China
| | - Yanqiu Li
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Youwei Sun
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Xingming Deng
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| | - Fu Wang
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, China.
| |
Collapse
|
35
|
Cheng YT, Yang CC, Shyur LF. Phytomedicine-Modulating oxidative stress and the tumor microenvironment for cancer therapy. Pharmacol Res 2016; 114:128-143. [PMID: 27794498 DOI: 10.1016/j.phrs.2016.10.022] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/21/2016] [Accepted: 10/24/2016] [Indexed: 12/18/2022]
Abstract
In spite of the current advances and achievements in systems biology and translational medicinal research, the current strategies for cancer therapy, such as radiotherapy, targeted therapy, immunotherapy and chemotherapy remain palliative or unsatisfactory due to tumor metastasis or recurrence after surgery/therapy, drug resistance, adverse side effects, and so on. Oxidative stress (OS) plays a critical role in chronic/acute inflammation, carcinogenesis, tumor progression, and tumor invasion/metastasis which is also attributed to the dynamic and complex properties and activities in the tumor microenvironment (TME). Re-educating or reprogramming tumor-associated stromal or immune cells in the TME provides an approach for restoring immune surveillance impaired by disease in cancer patients to increase overall survival and reduce drug resistance. Herbal medicines or plant-derived natural products have historically been a major source of anti-cancer drugs. Delving into the lore of herbal medicine may uncover new leads for anti-cancer drugs. Phytomedicines have been widely documented to directly or indirectly target multiple signaling pathways and networks in cancer cells. A combination of anti-cancer drugs and polypharmacological plant-derived extracts or compounds may offer a significant advantage in sensitizing the efficacy of monotherapy and overcoming drug-induced resistance in cancer patients. This review introduces several phytochemicals and phytoextracts derived from medicinal plants or dietary vegetables that have been studied for their efficacy in preclinical cancer models. We address the underlying modes of action of induction of OS and deregulation of TME-associated stromal cells, mediators and signaling pathways, and reference the related clinical investigations that look at the single or combination use of phytochemicals and phytoextracts to sensitize anti-cancer drug effects and/or overcome drug resistance.
Collapse
Affiliation(s)
- Yu-Ting Cheng
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan; Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
| | - Chun-Chih Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taiwan
| | - Lie-Fen Shyur
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan; Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan; Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taiwan; Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
36
|
The anti-proliferative and anti-androgenic activity of different pomegranate accessions. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.08.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
37
|
Cranberries and Cancer: An Update of Preclinical Studies Evaluating the Cancer Inhibitory Potential of Cranberry and Cranberry Derived Constituents. Antioxidants (Basel) 2016; 5:antiox5030027. [PMID: 27548236 PMCID: PMC5039576 DOI: 10.3390/antiox5030027] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/11/2016] [Accepted: 08/15/2016] [Indexed: 12/29/2022] Open
Abstract
Cranberries are rich in bioactive constituents reported to influence a variety of health benefits, ranging from improved immune function and decreased infections to reduced cardiovascular disease and more recently cancer inhibition. A review of cranberry research targeting cancer revealed positive effects of cranberries or cranberry derived constituents against 17 different cancers utilizing a variety of in vitro techniques, whereas in vivo studies supported the inhibitory action of cranberries toward cancers of the esophagus, stomach, colon, bladder, prostate, glioblastoma and lymphoma. Mechanisms of cranberry-linked cancer inhibition include cellular death induction via apoptosis, necrosis and autophagy; reduction of cellular proliferation; alterations in reactive oxygen species; and modification of cytokine and signal transduction pathways. Given the emerging positive preclinical effects of cranberries, future clinical directions targeting cancer or premalignancy in high risk cohorts should be considered.
Collapse
|
38
|
Borowska S, Brzóska MM. Chokeberries (Aronia melanocarpa
) and Their Products as a Possible Means for the Prevention and Treatment of Noncommunicable Diseases and Unfavorable Health Effects Due to Exposure to Xenobiotics. Compr Rev Food Sci Food Saf 2016; 15:982-1017. [DOI: 10.1111/1541-4337.12221] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/15/2016] [Accepted: 06/21/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Sylwia Borowska
- Borowska and Brzóska are with Dept. of Toxicology; Medical Univ. of Bialystok; Bialystok Poland
| | - Malgorzata M. Brzóska
- Borowska and Brzóska are with Dept. of Toxicology; Medical Univ. of Bialystok; Bialystok Poland
| |
Collapse
|
39
|
Al-Asmari AK, Khan AQ, Al-Masri N. Mitigation of 5-fluorouracil-induced liver damage in rats by vitamin C via targeting redox-sensitive transcription factors. Hum Exp Toxicol 2016; 35:1203-1213. [PMID: 26921358 DOI: 10.1177/0960327115626583] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adverse complications associated with antineoplastic drug-based cancer therapy are the major clinical drawbacks. Oxidative stress and inflammation play a major role in the damage due to cancer therapy. In the current study, we investigated the modulatory effect of vitamin C (Vit. C) on liver toxicity induced by 5-fluorouracil (5-FU) in rats. Animals were divided into four groups. Animals in group I received vehicle. Oral gavage of Vit. C (500 mg kg-1 body weight (b.wt.)) was given to the animals in group III and group IV. 5-FU (150 mg kg-1 b.wt.) was injected intraperitoneally to the animals in group II and group III. Findings of the present study revealed that oral administration of Vit. C significantly ameliorated the level of lipid peroxidation and the activity of myeloperoxidase. Vit. C administration markedly reduced the activation of nuclear factor κB and expression of cyclooxygenase 2, whereas nuclear translocation of nuclear factor erythroid 2-related factor 2 was increased. Hepatic histopathological analyses further supported the protective effect of Vit. C. Findings of the current study demonstrate that the toxic free radicals and inflammatory mediators generated due to chemotherapy play a critical role in 5-FU-induced hepatic damage. Attenuating action of Vit. C may be due to the modulation of redox-sensitive transcription factors and associated target molecules.
Collapse
Affiliation(s)
- A K Al-Asmari
- 1 Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - A Q Khan
- 1 Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - N Al-Masri
- 2 Department of Hepatology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
40
|
Liu C, Dai L, Liu Y, Rong L, Dou D, Sun Y, Ma L. Antiproliferative Activity of Triterpene Glycoside Nutrient from Monk Fruit in Colorectal Cancer and Throat Cancer. Nutrients 2016; 8:E360. [PMID: 27304964 PMCID: PMC4924201 DOI: 10.3390/nu8060360] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/27/2016] [Accepted: 06/03/2016] [Indexed: 01/10/2023] Open
Abstract
Colorectal cancer and throat cancer are the world's most prevalent neoplastic diseases, and a serious threat to human health. Plant triterpene glycosides have demonstrated antitumor activity. In this study, we investigated potential anticancer effects of mogroside IVe, a triterpenoid glycoside from monk fruit, using in vitro and in vivo models of colorectal and laryngeal cancer. The effects of mogroside IVe on the proliferation of colorectal cancer HT29 cells and throat cancer Hep-2 cells were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and the expression levels of p53, phosphorylated ERK1/2, and MMP-9 were analyzed by western blotting and immunohistochemistry. The results indicated that mogroside IVe inhibited, in a dose-dependent manner, the proliferation of HT29 and Hep-2 cells in culture and in xenografted mice, which was accompanied by the upregulation of tumor suppressor p53, and downregulation of matrix metallopeptidase 9 (MMP-9) and phosphorylated extracellular signal-regulated kinases (ERK)1/2. This study revealed the suppressive activity of mogroside IVe towards colorectal and throat cancers and identified the underlying mechanisms, suggesting that mogroside IVe may be potentially used as a biologically-active phytochemical supplement for treating colorectal and throat cancers.
Collapse
Affiliation(s)
- Can Liu
- Key Laboratory of Urban Agriculture (North) of Ministry of Agriculture, Beijing University of Agriculture, Beijing 102206, China.
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China.
- Beijing Collaborative Innovation Center for Eco-Environmental Improvement with Forestry and Fruit Trees, Beijing 102206, China.
| | - Longhai Dai
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China.
| | - Yueping Liu
- Key Laboratory of Urban Agriculture (North) of Ministry of Agriculture, Beijing University of Agriculture, Beijing 102206, China.
- Beijing Collaborative Innovation Center for Eco-Environmental Improvement with Forestry and Fruit Trees, Beijing 102206, China.
| | - Long Rong
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Dequan Dou
- Key Laboratory of Urban Agriculture (North) of Ministry of Agriculture, Beijing University of Agriculture, Beijing 102206, China.
| | - Yuanxia Sun
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China.
| | - Lanqing Ma
- Key Laboratory of Urban Agriculture (North) of Ministry of Agriculture, Beijing University of Agriculture, Beijing 102206, China.
- Beijing Collaborative Innovation Center for Eco-Environmental Improvement with Forestry and Fruit Trees, Beijing 102206, China.
| |
Collapse
|