1
|
Shadnoush M, Momenan M, Seidel V, Tierling S, Fatemi N, Nazemalhosseini-Mojarad E, Norooz MT, Cheraghpour M. A comprehensive update on the potential of curcumin to enhance chemosensitivity in colorectal cancer. Pharmacol Rep 2025; 77:103-123. [PMID: 39304638 DOI: 10.1007/s43440-024-00652-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Colorectal cancer (CRC) is one of the most common cancers and a major cause of cancer-related mortality worldwide. The efficacy of chemotherapy agents in CRC treatment is often limited due to toxic side effects, heterogeneity of cancer cells, and the possibility of chemoresistance which promotes cancer cell survival through several mechanisms. Combining chemotherapy agents with natural compounds like curcumin, a polyphenol compound from the Curcuma longa plant, has been reported to overcome chemoresistance and increase the sensitivity of cancer cells to chemotherapeutics. Curcumin, alone or in combination with chemotherapy agents, has been demonstrated to prevent chemoresistance by modulating various signaling pathways, reducing the expression of drug resistance-related genes. The purpose of this article is to provide a comprehensive update on studies that have investigated the ability of curcumin to enhance the efficacy of chemotherapy agents used in CRC. It is hoped that it can serve as a template for future research on the efficacy of curcumin, or other natural compounds, combined with chemotherapy agents to maximize the effectiveness of therapy and reduce the side effects that occur in CRC or other cancers.
Collapse
Affiliation(s)
- Mahdi Shadnoush
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.Box, Tehran, 16635-148, Iran
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnaz Momenan
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Sascha Tierling
- Department of Genetics/Epigenetics, Faculty NT, Life Sciences, Saarland University, Saarbrücken, Germany
| | - Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.Box, Tehran, 16635-148, Iran
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Tayefeh Norooz
- General Surgery Department, Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Makan Cheraghpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.Box, Tehran, 16635-148, Iran.
| |
Collapse
|
2
|
Nguyen MH, Le NTH, Nguyen BQH, Nguyen MTT, Do TNV, Le TH, Nguyen VT, Yen CH. In vitro and in silico hybrid approach to unveil triterpenoids from Helicteres hirsuta leaves as potential compounds for inhibiting Nrf2. RSC Adv 2025; 15:1915-1923. [PMID: 39839230 PMCID: PMC11749225 DOI: 10.1039/d4ra07646j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/28/2024] [Indexed: 01/23/2025] Open
Abstract
Cancer is a leading global health concern, with over 20 million new cases and 9.7 million deaths reported in 2022. Chemotherapy remains a widely used treatment, but drug resistance, which affects up to 90% of treatment outcomes, significantly hampers its effectiveness. The transcription factor Nrf2, which is crucial for cellular defense against oxidative stress, plays a dual role in cancer treatment. Although Nrf2 activation can suppress early carcinogenesis, its overexpression in cancer cells contributes to drug resistance, resulting in poor patient outcomes. Thus, inhibiting Nrf2 has emerged as a promising strategy for overcoming cancer drug resistance. Natural compounds such as luteolin and brusatol have shown potential in inhibiting Nrf2, although with limitations. This study isolates and characterizes seven triterpenoids from the n-hexane sub-fraction of Helicteres hirsuta, a plant traditionally used for medicinal purposes, to evaluate their ability to modulate Nrf2 activity in Huh7 cancer and HaCaT normal cells. Additionally, molecular docking and dynamic simulations were utilized to assess the binding potential of these compounds to the PI3Kα receptor, which regulates downstream signaling pathways, thereby suppressing Nrf2 activity in cancer cells. Our findings provide insights into new strategies seeking triterpenoids as promising structures to reverse chemoresistance by regulating Nrf2. The results also reveal the potential of 3β-O-trans-caffeoylbetulinic acid from H. hirsuta leaves as the unprecedented compound inhibiting Nrf2 activity, with an IC50 of 74.5 μg mL-1 in Huh7 cancer cells.
Collapse
Affiliation(s)
- Minh Hien Nguyen
- University of Health Sciences, Vietnam National University Ho Chi Minh City YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward Di An City Binh Duong Province 75308 Vietnam (+84) 373 696 894
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
| | - Nguyen Thien Han Le
- University of Health Sciences, Vietnam National University Ho Chi Minh City YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward Di An City Binh Duong Province 75308 Vietnam (+84) 373 696 894
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
| | - Bui Quoc Huy Nguyen
- The University of Danang - VN-UK Institute for Research and Executive Education 41 Le Duan Street, Hai Chau 1 Ward, Hai Chau District Danang City 50000 Vietnam
| | - Mai Thanh Thi Nguyen
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
- Faculty of Chemistry, University of Science Ho Chi Minh City Vietnam
- Research Lab for Drug Discovery and Development, University of Science Ho Chi Minh City Vietnam
| | - Truong Nhat Van Do
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
- Faculty of Chemistry, University of Science Ho Chi Minh City Vietnam
- Research Lab for Drug Discovery and Development, University of Science Ho Chi Minh City Vietnam
| | - Tho Huu Le
- Vietnam National University Ho Chi Minh City Quarter 6, Linh Trung Ward, Thu Duc District Ho Chi Minh City 70000 Vietnam
- Faculty of Chemistry, University of Science Ho Chi Minh City Vietnam
- Research Lab for Drug Discovery and Development, University of Science Ho Chi Minh City Vietnam
| | - Vu Thanh Nguyen
- Division of Aquacultural Biotechnology, Biotechnology Center of Ho Chi Minh City 2374, Highway 1, Quarter 2, Trung My Tay Ward, District 12 Ho Chi Minh City 70000 Vietnam
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University No. 100, Shih-Chuan 1st Road, Sanmin District Kaohsiung City 80708 Taiwan
- National Natural Product Libraries and High-Throughput Screening Core Facility, Kaohsiung Medical University No. 100, Shih-Chuan 1st Road, Sanmin District Kaohsiung City 80708 Taiwan
| |
Collapse
|
3
|
Yilmaz YB, Güngör T, Dönmez S, Atalay HN, Siyah P, Durdağı S, Ay M, Boyunegmez Tumer T. Synthesis, in silico and bio-evaluation studies of new isothiocyanate derivatives with respect to COX inhibition and H 2S release profiles. RSC Med Chem 2024:d4md00495g. [PMID: 39507615 PMCID: PMC11536671 DOI: 10.1039/d4md00495g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
The development of H2S-donating derivatives of non-steroidal anti-inflammatory drugs (NSAIDs) is considered important to reduce or overcome their gastrointestinal side effects. Sulforaphane, one of the most extensively studied isothiocyanates (ITCs), effectively releases H2S at a slow rate. Thus, we rationally designed, synthesized, and characterized new ITC derivatives (I1-3 and I1a-e) inspired by the natural compound sulforaphane. The anti-inflammatory properties of these compounds were evaluated by their inhibitory activities against cyclooxygenase targets COX-1 and COX-2. Additionally, the cytotoxicity of the compounds was tested using the MTT assay on LPS-induced RAW 264.7 cells, revealing no cytotoxic effects at low doses. Notably, compounds I1 and fluorine-containing ester derivative I1c emerged as the most potent and selective COX-2 inhibitors, with selectivity indexes of 2611.5 and 2582.4, respectively. The H2S-releasing capacities of ITC derivatives were investigated and compared with that of sulforaphane, showing that while compounds I1-3 exhibit slow and similar H2S release to sulforaphane, the release from compounds I1a-e was not as pronounced as that of the standard. Physics-based molecular modeling studies including molecular docking and molecular dynamics (MD) simulations, binding free energy calculations and absorption, distribution, metabolism, and excretion (ADME) analyses were also conducted. MD simulations analysis underscored the crucial amino acids such as Tyr385, Trp387, Phe518, Val523, and Ser530 in the interactions between I1c hit compound and COX-2. The combined in silico and in vitro findings suggest that compounds I1 and I1c are promising NSAID candidates against selective COX-2 inhibition.
Collapse
Affiliation(s)
- Yakup Berkay Yilmaz
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Tuğba Güngör
- Natural Products and Drug Research Laboratory, Department of Chemistry, Faculty of Science, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Serhat Dönmez
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Hazal Nazlıcan Atalay
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Pınar Siyah
- Department of Biochemistry, School of Pharmacy, Bahçeşehir University Istanbul Türkiye
| | - Serdar Durdağı
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University Istanbul Türkiye
- Molecular Therapy Lab, Department of Pharmaceutical Chemistry, School of Pharmacy, Bahçeşehir University Istanbul Türkiye
- Lab for Innovative Drugs (Lab4IND), Computational Drug Design Center (HITMER), Bahçeşehir University Istanbul Türkiye
| | - Mehmet Ay
- Natural Products and Drug Research Laboratory, Department of Chemistry, Faculty of Science, Canakkale Onsekiz Mart University Canakkale Türkiye
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Science, Canakkale Onsekiz Mart University Canakkale Türkiye
| |
Collapse
|
4
|
Oncu S, Becit-Kizilkaya M, Sen S, Ugur-Kaplan AB, Cetin M, Celik S. Daidzein nanosuspension in combination with cisplatin to enhance therapeutic efficacy against A549 non-small lung cancer cells: an in vitro evaluation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4871-4881. [PMID: 38159158 DOI: 10.1007/s00210-023-02924-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Lung cancer is the most common cause of cancer-related mortality, chemo-resistance, and toxicity limit treatment. The focus is on innovative combined phytotherapy to improve treatment outcomes. Our aim was to investigate the potential effects of daidzein nanosuspension (DZ-NS) and its combination with cisplatin (CIS) on A549 non-small lung cancer cells. Cytotoxicity was investigated using MTT and Chou-Talalay methods. Oxidative, apoptotic, and inflammatory markers were analyzed by ELISA and qRT-PCR. The IC50 value for DZ-NS was 25.23 µM for 24 h and was lower than pure DZ (IC50 = 835 µM for pure DZ). DZ-NS (at IC50x2 and IC50 values) showed synergistic cytotoxicity with CIS. The cells treated with DZ-NS had low TOS and OSI levels. However, DZ-NS failed to regulate Cas3 and TGF-β1 activation in A549 cells. MMP-9 gene expression was significantly suppressed in DZ-NS-treated cells, especially in combination therapy. DZ represents a potential combination option for the treatment of lung cancer, and its poor toxicokinetic properties limit its clinical use. To overcome these limitations, the effects of the nanosuspension formulation were tested. DZ-NS showed a cytotoxic effect on A549 cells and optimized the therapeutic effect of CIS. This in vitro synergistic effect was mediated by suppression of MMP-9 and not by oxidative stress or Cas3-activated apoptosis. This study provides the basis for an in vivo and clinical trial of DZ-NS with concurrent chemotherapy.
Collapse
Affiliation(s)
- Seyma Oncu
- Department of Medical Pharmacology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Merve Becit-Kizilkaya
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Afyonkarahisar Health Sciences University, Afyonkarahisar, 03030, Turkey.
| | - Serkan Sen
- Department of Medical Laboratory Techniques, Ataturk Vocational School of Health Services, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Afife Busra Ugur-Kaplan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - Meltem Cetin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - Sefa Celik
- Department of Medical Biochemistry, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| |
Collapse
|
5
|
Liu P, Zhang B, Li Y, Yuan Q. Potential mechanisms of cancer prevention and treatment by sulforaphane, a natural small molecule compound of plant-derived. Mol Med 2024; 30:94. [PMID: 38902597 PMCID: PMC11191161 DOI: 10.1186/s10020-024-00842-7] [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: 12/28/2023] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Despite recent advances in tumor diagnosis and treatment technologies, the number of cancer cases and deaths worldwide continues to increase yearly, creating an urgent need to find new methods to prevent or treat cancer. Sulforaphane (SFN), as a member of the isothiocyanates (ITCs) family, which is the hydrolysis product of glucosinolates (GLs), has been shown to have significant preventive and therapeutic cancer effects in different human cancers. Early studies have shown that SFN scavenges oxygen radicals by increasing cellular defenses against oxidative damage, mainly through the induction of phase II detoxification enzymes by nuclear factor erythroid 2-related factor 2 (Nrf2). More and more studies have shown that the anticancer mechanism of SFN also includes induction of apoptotic pathway in tumor cells, inhibition of cell cycle progression, and suppression of tumor stem cells. Therefore, the application of SFN is expected to be a necessary new approach to treating cancer. In this paper, we review the multiple molecular mechanisms of SFN in cancer prevention and treatment in recent years, which can provide a new vision for cancer treatment.
Collapse
Affiliation(s)
- Pengtao Liu
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Bo Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Yuanqiang Li
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China.
| |
Collapse
|
6
|
Sailo BL, Liu L, Chauhan S, Girisa S, Hegde M, Liang L, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Harnessing Sulforaphane Potential as a Chemosensitizing Agent: A Comprehensive Review. Cancers (Basel) 2024; 16:244. [PMID: 38254735 PMCID: PMC10814109 DOI: 10.3390/cancers16020244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Recent advances in oncological research have highlighted the potential of naturally derived compounds in cancer prevention and treatment. Notably, sulforaphane (SFN), an isothiocyanate derived from cruciferous vegetables including broccoli and cabbage, has exhibited potent chemosensitizing capabilities across diverse cancer types of bone, brain, breast, lung, skin, etc. Chemosensitization refers to the enhancement of cancer cell sensitivity to chemotherapy agents, counteracting the chemoresistance often developed by tumor cells. Mechanistically, SFN orchestrates this sensitization by modulating an array of cellular signaling pathways (e.g., Akt/mTOR, NF-κB, Wnt/β-catenin), and regulating the expression and activity of pivotal genes, proteins, and enzymes (e.g., p53, p21, survivin, Bcl-2, caspases). When combined with conventional chemotherapeutic agents, SFN synergistically inhibits cancer cell proliferation, invasion, migration, and metastasis while potentiating drug-induced apoptosis. This positions SFN as a potential adjunct in cancer therapy to augment the efficacy of standard treatments. Ongoing preclinical and clinical investigations aim to further delineate the therapeutic potential of SFN in oncology. This review illuminates the multifaceted role of this phytochemical, emphasizing its potential to enhance the therapeutic efficacy of anti-cancer agents, suggesting its prospective contributions to cancer chemosensitization and management.
Collapse
Affiliation(s)
- Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| | - Le Liu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen 518001, China;
| | - Suravi Chauhan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China;
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia;
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia;
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| |
Collapse
|
7
|
Wang G, Zhao R, Zhang X, Zheng Y, Xie F, Jiang Y, Lv G, Long D, Sun C, Bao Y, Qi S, Liu X, Zhang Q, Yang X. EGFR/MAPK signaling pathway acts as a potential therapeutic target for sulforaphane-rescued heart tube malformation induced by various concentrations of PhIP exposure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155270. [PMID: 38096717 DOI: 10.1016/j.phymed.2023.155270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/14/2023] [Accepted: 12/07/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND 2-Amino-1-methyl-6-phenylimidazo [4,5-b] pyrimidine (PhIP) is a known carcinogen generated mainly from cooking meat and environmental pollutants. It is worth exploring the potential of natural small-molecule drugs to protect against adverse effects on embryonic development. PURPOSE In this study, we investigated the potential toxicological effects of PhIP on embryonic heart tube formation and the effect of Sulforaphane (SFN) administration on the anti-toxicological effects of PhIP on embryonic cardiogenesis. STUDY DESIGN AND METHODS First, the chicken embryo model was used to investigate the different phenotypes of embryonic heart tubes induced by various concentrations of PhIP exposure. We also proved that SFN rescues PhIP-induced embryonic heart tube malformation. Second, immunofluorescence, western blot, Polymerase Chain Reaction (PCR) and flow cytometry experiments were employed to explore the mechanisms by which SFN protects cardiac cells from oxidative damage in the presence of PhIP. We used RNA-seq analysis, molecular docking, in situ hybridization, cellular thermal shift assay and solution nuclear magnetic resonance spectroscopy to explore whether SFN protects cardiogenesis through the EGFR/MAPK signaling pathway. RESULTS The study showed that PhIP might dose-dependently interfere with the C-looping heart tube (mild) or the fusion of a pair of bilateral endocardial tubes (severe) in chick embryos, while SFN administration prevented cardiac cells from oxidative damage in the presence of high-level PhIP. Furthermore, we found that excessive reactive oxygen species (ROS) production and subsequent apoptosis were not the principal mechanisms by which low-level PhIP induced malformation of heart tubes. This is due to PhIP-disturbed Mitogen-activated protein kinase (MAPK) signaling pathway could be corrected by SFN administration. CONCLUSIONS This study provided novel insight that PhIP exposure could increase the risk of abnormalities in early cardiogenesis and that SFN could partially rescue various concentrations of PhIP-induced abnormal heart tube formation by targeting EGFR and mediating EGFR/MAPK signaling pathways.
Collapse
Affiliation(s)
- Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China.
| | - Ran Zhao
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Xinxia Zhang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Ying Zheng
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Feiling Xie
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Yu Jiang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Guohua Lv
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Denglu Long
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Chengyang Sun
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7UQ, United Kingdom
| | - Shuangyu Qi
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Xinyue Liu
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Qihao Zhang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China; Clinical Research Center, Clifford Hospital, Guangzhou 511495, China.
| |
Collapse
|
8
|
Javed J, Anjum I, Najm S, Ali N, Nasir Hayat Malik M, Jahan S, Dawoud TM, Nafidi HA, Bourhia M. Uroprotective Potential of Campesterol in Cyclophosphamide Induced Interstitial Cystitis; Molecular Docking Studies. Chem Biodivers 2023; 20:e202301534. [PMID: 37984454 DOI: 10.1002/cbdv.202301534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
Cyclophosphamide (CYP) is commonly used to treat cancer of the ovaries, breast, lymph, and blood system and produces interstitial cystitis (IC) via its urotoxic metabolite: i. e., acrolein. The present study was aimed to investigate the uroprotective effect of campesterol (a steroidal phytochemical) in cyclophosphamide induced IC. IC was induced by CYP (150 mg/kg, i. p.) in rats. The Enzyme linked immunosorbent assays for oxidative stress markers and Polymerase Chain Reaction (PCR) for inflammatory cytokines were carried out. The Tissue Organ Bath Technique was used for the evaluation of the spasmolytic effect of campesterol. Different pharmacological antagonists have been used to explore the mechanism of action of campesterol. Treatment with campesterol (70 mg/kg) reduced nociception (55 %), edema (67 %), hemorrhage (67 %), and protein leakage significantly (94 %). The antioxidant activity of campesterol was exhibited by a fall in MDA, NO, and an elevation in SOD, CAT, and GPX levels. Campesterol presented anti-inflammatory potential by decreasing IL-1, TNF-α, and TGF-β expression levels. Histologically, it preserved urothelium from the deleterious effect of CYP. Campesterol showed a spasmolytic effect by reducing bladder overactivity that was dependent on muscarinic receptors, voltage-gated calcium and KATP channels, and cyclo-oxygenase pathways. In silico studies confirmed the biochemical findings. The findings suggest that campesterol could be valorized as a possible therapeutic agent against cyclophosphamide-induced interstitial cystitis.
Collapse
Affiliation(s)
- Joham Javed
- Faculty of Pharmacy, The University of Lahore, Lahore, 55150, Pakistan
| | - Irfan Anjum
- Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, 44000, Pakistan
| | - Saima Najm
- Lahore College of Pharmaceutical Sciences, Department of Pharmacy, Lahore, 55150, Pakistan
| | - Naila Ali
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 55150, Pakistan
| | | | - Shah Jahan
- Department of Immunology, University of Health Sciences, Lahore, 55150, Pakistan
| | - Turki M Dawoud
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. BOX 2455, Riyadh, 11451, Saudi Arabia
| | - Hiba-Allah Nafidi
- Department of Food Science, Faculty of Agricultural and Food Sciences, Laval University, 2325, Quebec City, QC G1 V0 A6, Canada
| | - Mohammed Bourhia
- Department of Chemistry and Biochemistry, Faculty of Medicine and Pharmacy, Ibn Zohr University, Laayoune, 70000, Morocco
| |
Collapse
|
9
|
Ulusan M, Bireller S, Ertugrul B, Kasarci G, Atas MN, Aydemir L, Ergen A, Cakmakoglu B. What if amoxicillin/clavulanic acid reduces the cisplatin anticancer impact on oral cancer treatment? JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2023; 124:101502. [PMID: 37192700 DOI: 10.1016/j.jormas.2023.101502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 05/18/2023]
Abstract
Antibiotics-chemotherapeutics combination have become on the table for many cancer treatments. For this reason, we thought that further progress and development of studies to support chemotherapeutic approaches with the use of antibiotics may be beneficial in the clinical field. Cell lines (SCC-15, HTB-41, and MRC-5) were treated with 5-100 μM/ml concentrations of cisplatin (cisp) and amoxicillin/clavulanic acid (amx/cla) with combination (amx/cla-cisp) and alone in three different incubation periods. The all-cells viability was examined with WST-1 and apoptotic activity of the drugs were investigated via cell death ELISA assay kit. The cytotoxic impact of the 100 μM amx/cla-cisp combination was found to be reduced by up to 21.8%, which was significant given that the cytotoxic effect of only cisplatin therapy was 86.1%. Because our findings demonstrated that solo amx/cla therapy have almost no impact on proliferation or death, we focused on the amx/cla-cisp combination effect. It was found that the amx/cla-cisp combination has reduced the apoptotic fragment when comparing with the solely cisp-treated cells. Due to amx/cla-cisp combination on both cells but significantly on SCC-15 recovered the sole cisplatin effect, we believe that there might be a second thought when prescribing antibiotics while treating cancer patients. Not only the antibiotic's type but also the cancer type might interact to lessen the chemotherapeutic agent's impact which is clinically a dilemma to focus on.
Collapse
Affiliation(s)
- Murat Ulusan
- Department of Otolaryngology, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sinem Bireller
- Department of Biochemistry, Faculty of Pharmacy, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - Baris Ertugrul
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Goksu Kasarci
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Merve Nur Atas
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Levent Aydemir
- Department of Otolaryngology, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Arzu Ergen
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Bedia Cakmakoglu
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
10
|
Wu S, Li L, Liang Q, Gao H, Tang T, Tang Y. A DFT study of sulforaphane adsorption on the group III nitrides (B12N12, Al12N12 and Ga12N12) nanocages. J Biomol Struct Dyn 2023; 42:12730-12741. [PMID: 37882329 DOI: 10.1080/07391102.2023.2272755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/04/2023] [Indexed: 10/27/2023]
Abstract
In this paper, the adsorption behavior of group III nitrides (B12N12, Al12N12, and Ga12N12) nanocages to sulforaphane (SF) anticancer medicine were studied by density functional theory (DFT). The adsorption energy, solvation energy, desorption time and related quantum molecular descriptors were calculated in neutral and acidic solutions. When the drugs were adsorbed to nanocages, the structure of nanocages and drugs changed after adsorption, indicating that the process was effective adsorption. The adsorption energy and solvation energy of the complexes created after adsorption were negative values, which indicated that the structure of complexes formed by adsorption were stable. According to charge decomposition analysis (CDA) and natural bonding orbitals (NBO), drugs act as charge donors and nanocages act as charge acceptors, so that the charge flows from drugs to nanocages. Thermodynamic calculations demonstrate that drugs adsorption on nanocages is a spontaneous exothermic process. The calculation of quantum molecular descriptors confirmed that drugs adsorption on nanocages increased the chemical reactivity and solubility of drugs, which facilitated its transfer in biological fluids. Both interaction region index (IRI) and topological analysis of atom in molecule (AIM) revealed Van Der Waals interaction between drugs and nanocages. Protonation studies demonstrated that acidic circumstances could improve the polarity of complexes, increase the solvation effect, and boost drugs release in target cancer cells. The results of this work indicate that X12N12(X = B, Al, Ga) nanocages can be used as the delivery vehicle of SF drug.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- ShiQuan Wu
- School of Physics, Guizhou University, Guiyang, China
| | - Li Li
- School of Physics, Guizhou University, Guiyang, China
| | - QiQi Liang
- School of Physics, Guizhou University, Guiyang, China
| | - HuaXu Gao
- School of Physics, Guizhou University, Guiyang, China
| | - TianYu Tang
- School of Physics, Guizhou University, Guiyang, China
| | - YanLin Tang
- School of Physics, Guizhou University, Guiyang, China
| |
Collapse
|
11
|
Monteiro-Alfredo T, dos Santos JM, Antunes KÁ, Cunha J, da Silva Baldivia D, Pires AS, Marques I, Abrantes AM, Botelho MF, Monteiro L, Gonçalves AC, Botelho WH, Paula de Araújo Boleti A, Cabral C, Oliveira PJ, Lucas dos Santos E, Matafome P, de Picoli Souza K. Acrocomia aculeata associated with doxorubicin: cardioprotection and anticancer activity. Front Pharmacol 2023; 14:1223933. [PMID: 37654604 PMCID: PMC10466431 DOI: 10.3389/fphar.2023.1223933] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/01/2023] [Indexed: 09/02/2023] Open
Abstract
Doxorubicin (Dox) is a chemotherapeutic agent widely used in the clinic, whose side effects include cardiotoxicity, associated with decreased antioxidant defenses and increased oxidative stress. The association of Dox with natural antioxidants can extend its use if not interfering with its pharmacological potential. In this study, we aimed to understand the effects and mechanisms of the aqueous extract of Acrocomia aculeata leaves (EA-Aa) in cancer cells and the co-treatment with Dox, in in vitro and in vivo models. It was found that EA-Aa showed a relevant decrease in the viability of cancer cells (K562 and MCF-7) and increased apoptosis and death. The Dox cytotoxic effect in co-treatment with EA-Aa was increased in cancer cells. The therapeutic association also promoted a change in cell death, leading to a higher rate of apoptosis compared to the Dox group, which induced necrosis. In addition, in non-cancer cells, EA-Aa enhanced red blood cell (RBC) redox state with lower hemolysis and malondialdehyde (MDA) content and had no in vitro nor in vivo toxicity. Furthermore, EA-Aa showed antioxidant protection against Dox-induced cytotoxicity in H9c2 cells (cardiomyoblast), partially mediated by the NRF2 pathway. In vivo, EA-Aa treatment showed a relevant decrease in MDA levels in the heart, kidney, and brain, evaluated in C57Bl/6 mice induced to cardiotoxicity by Dox. Together, our results proved the effectiveness of EA-Aa in potentiating Dox anticancer effects, with antioxidant and cardioprotective activity, suggesting EA-Aa as a potential Dox pharmacological adjuvant.
Collapse
Affiliation(s)
- Tamaeh Monteiro-Alfredo
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
- Faculty of Medicine, Institute of Physiology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Jéssica Maurino dos Santos
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Kátia Ávila Antunes
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Janielle Cunha
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Debora da Silva Baldivia
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Ana Salomé Pires
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Inês Marques
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Ana Margarida Abrantes
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Maria Filomena Botelho
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Lúcia Monteiro
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
| | - Wellington Henrique Botelho
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Ana Paula de Araújo Boleti
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Célia Cabral
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Paulo J. Oliveira
- CNC—Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Edson Lucas dos Santos
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Paulo Matafome
- Faculty of Medicine, Institute of Physiology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Department of Complementary Sciences, Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Kely de Picoli Souza
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| |
Collapse
|
12
|
Astrain-Redin N, Raza A, Encío I, Sharma AK, Plano D, Sanmartín C. Novel Acylselenourea Derivatives: Dual Molecules with Anticancer and Radical Scavenging Activity. Antioxidants (Basel) 2023; 12:1331. [PMID: 37507871 PMCID: PMC10376326 DOI: 10.3390/antiox12071331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Oxidative stress surrounding cancer cells provides them with certain growth and survival advantages necessary for disease progression. In this context, Se-containing molecules have gained attention due to their anticancer and antioxidant activity. In our previous work, we synthesized a library of 39 selenoesters containing functional groups commonly present in natural products (NP), which showed potent anticancer activity, but did not demonstrate high radical scavenger activity. Thus, 20 novel Se derivatives resembling NP have been synthesized presenting acylselenourea functionality in their structures. Radical scavenger activity was tested using DPPH assay and in vitro protective effects against ROS-induced cell death caused by H2O2. Additionally, antiproliferative activity was evaluated in prostate, colon, lung, and breast cancer cell lines, along with their ability to induce apoptosis. Compounds 1.I and 5.I showed potent cytotoxicity against the tested cancer cell lines, along with high selectivity indexes and induction of caspase-mediated apoptosis. These compounds exhibited potent and concentration-dependent radical scavenging activity achieving DPPH inhibition similar to ascorbic acid and trolox. To conclude, we have demonstrated that the introduction of Se in the form of acylselenourea into small molecules provides strong radical scavengers in vitro and antiproliferative activity, which may lead to the development of promising dual compounds.
Collapse
Affiliation(s)
- Nora Astrain-Redin
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Asif Raza
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Ignacio Encío
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Avda. Barañain s/n, 31008 Pamplona, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Daniel Plano
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain
| | - Carmen Sanmartín
- Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea, 3, 31008 Pamplona, Spain
| |
Collapse
|
13
|
Peng X, Zhang X, Wang C, Olatunji OJ. Protective effects of asperuloside against cyclophosphamide-induced urotoxicity and hematotoxicity in rats. OPEN CHEM 2022. [DOI: 10.1515/chem-2022-0234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
Cyclophosphamide (CP) is a highly efficacious chemotherapy drug for treating cancers and autoimmune disorders, but it is also notable for its deleterious side effects including urotoxicity in cancer patients, which has been extensively linked to CP-induced oxidative/inflammatory cascades. Herein, we investigated the protective effects of asperuloside (ASP) against CP-induced urotoxicity. Rats received oral administration of ASP (20 and 40 mg/kg bw/day) for 35 days and were injected with weekly CP (100 mg/kg bw, i.p.) for 4 weeks to induce acute bladder toxicity. CP acutely altered haematological parameters and significantly reduced body weight gain, bladder glutathione peroxidase, reduced glutathione, catalase, and superoxide dismutase activities. Furthermore, CP caused an upward surge in bladder malondialdehyde, nuclear factor-kappa B, tumour necrosis factor-α, interleukin-1β, and interleukin 6 concentrations. ASP supplementation ameliorated CP-induced haematological derangement and bladder urotoxicity through the restoration of oxidative and inflammatory parameters in CP-treated rats. These findings suggested that ASP could be valorised as a possible therapeutic agent against chemotherapy-related toxicities as well as oxidative damage disorders.
Collapse
Affiliation(s)
- Xiaozhuang Peng
- Emergency Intensive Care Unit, Wuhu Second Peoples Hospital , Wuhu City , 241001, Anhui , China
| | - Xiaomin Zhang
- Emergency Intensive Care Unit, Wuhu Second Peoples Hospital , Wuhu City , 241001, Anhui , China
| | - Chen Wang
- Emergency Intensive Care Unit, Wuhu Second Peoples Hospital , Wuhu City , 241001, Anhui , China
| | - Opeyemi Joshua Olatunji
- Traditional Thai Medical Research and Innovation Center, Faculty of Traditional Thai Medicine, Prince of Songkla University , Hat Yai , 90110 , Thailand
| |
Collapse
|
14
|
Ghasemi H, Jamshidi A, Ghatee MA, Mazhab-Jafari K, Khorasani M, Rahmati M, Mohammadi S. PPARγ activation by pioglitazone enhances the anti-proliferative effects of doxorubicin on pro-monocytic THP-1 leukemia cells via inducing apoptosis and G2/M cell cycle arrest. J Recept Signal Transduct Res 2022; 42:429-438. [PMID: 34645362 DOI: 10.1080/10799893.2021.1988972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE Doxorubicin (DOX) is a common chemotherapeutic agent, with toxic side effects, and chemoresistance. Combination chemotherapy is a successful approach to overcome these limitations. Here, we investigated the effects of pioglitazone (PGZ), a PPARγ agonist, and/or DOX on the viability, cell cycle, apoptosis on THP-1 cells and normal human monocytes (NHMs). METHODS MTT assay was used to evaluate the cytotoxicity of DOX and/or PGZ. Cell cycle progression and apoptosis induction were examined by PI or Annexin V-PI double staining, and analyzed by flow cytometry. Quantitative RT-PCR was used to evaluate the changes in the mRNA expression of cell cycle progression or apoptosis-associated genes including P27, P21, CDK2, P53, BCL2 and FasR. RESULTS DOX, PGZ and DOX + PGZ exerted their cytotoxic effects in a dose- and time-dependent manner with low toxicity on NHMs. The cell growth inhibitory effects of DOX were in association with G2/M arrest, while PGZ executed S phase arrest. PGZ treatment enhanced G2/M among DOX-treated combinations with moderate elevation in the S phase. DOX, PGZ and combined treatments induced apoptosis (mostly late phase) in a dose-dependent manner. All treatments resulted in the significant overexpression of p21, p27, p53 and FasR genes and downregulation of CDK2. DOX + PGZ combined treatments exhibited the most significant changes in mRNA expression. CONCLUSION We demonstrated that the antiproliferative, cell cycle regulation and apoptosis-inducing capacity of DOX was enhanced by PGZ in THP-1 leukemia cells in a dose-dependent manner. Therefore, the combination of DOX + PGZ could be used as a novel combination to target AML.
Collapse
Affiliation(s)
- Hassan Ghasemi
- Department of Clinical Biochemistry, Abadan Faculty of Medical Sciences, Abadan, Iran
| | - Ali Jamshidi
- Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Mohammad Amin Ghatee
- Department of Medical Parasitology and Mycology, School of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Komeil Mazhab-Jafari
- Department of Laboratory Sciences, Abadan Faculty of Medical Sciences, Abadan, Iran
| | - Milad Khorasani
- Department of Clinical Biochemistry, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Mina Rahmati
- Metabolic Disorders Research Center, Department of Biochemistry, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Saeed Mohammadi
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
15
|
Famurewa AC, Mukherjee AG, Wanjari UR, Sukumar A, Murali R, Renu K, Vellingiri B, Dey A, Valsala Gopalakrishnan A. Repurposing FDA-approved drugs against the toxicity of platinum-based anticancer drugs. Life Sci 2022; 305:120789. [PMID: 35817170 DOI: 10.1016/j.lfs.2022.120789] [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: 05/24/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022]
Abstract
Platinum-based anticancer drugs (PADs), mainly cisplatin, carboplatin, and oxaliplatin, are widely used efficacious long-standing anticancer agents for treating several cancer types. However, clinicians worry about PAD chemotherapy and its induction of severe non-targeted organ toxicity. Compelling evidence has shown that toxicity of PAD on delicate body organs is associated with free radical generation, DNA impairment, endocrine and mitochondrial dysfunctions, oxidative inflammation, apoptosis, endoplasmic reticulum stress, and activation of regulator signaling proteins, cell cycle arrest, apoptosis, and pathways. The emerging trend is the repurposing of FDA-approved non-anticancer drugs (FNDs) for combating the side effects toxicity of PADs. Thus, this review chronicled the mechanistic preventive and therapeutic effects of FNDs against PAD organ toxicity in preclinical studies. FNDs are potential clinical drugs for the modulation of toxicity complications associated with PAD chemotherapy. Therefore, FNDs may be suggested as non-natural agent inhibitors of unpalatable side effects of PADs.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike lkwo, Nigeria.
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Aarthi Sukumar
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
16
|
Zaher S, Soliman ME, Elsabahy M, Hathout RM. Sesamol Loaded Albumin Nanoparticles: A Boosted Protective Property in Animal Models of Oxidative Stress. Pharmaceuticals (Basel) 2022; 15:ph15060733. [PMID: 35745652 PMCID: PMC9228363 DOI: 10.3390/ph15060733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 02/07/2023] Open
Abstract
The current study evaluated the ability of sesamol-loaded albumin nanoparticles to impart protection against oxidative stress induced by anthracyclines in comparison to the free drug. Albumin nanoparticles were prepared via the desolvation technique and then freeze-dried with the cryoprotectant, trehalose. Albumin concentration, pH, and type of desolvating agent were assessed as determining factors for successful albumin nanoparticle fabrication. The optimal nanoparticles were spherical in shape, and they had an average particle diameter of 127.24 ± 2.12 nm with a sesamol payload of 96.89 ± 2.4 μg/mg. The drug cellular protection was tested on rat hepatocytes pretreated with 1 µM doxorubicin, which showed a 1.2-fold higher protective activity than the free sesamol. In a pharmacokinetic study, the loading of a drug onto nanoparticles resulted in a longer half-life and mean residence time, as compared to the free drug. Furthermore, in vivo efficacy and biochemical assessment of lipid peroxidation, cardiac biomarkers, and liver enzymes were significantly ameliorated after administration of the sesamol-loaded albumin nanoparticles. The biochemical assessments were also corroborated with the histopathological examination data. Sesamol-loaded albumin nanoparticles, prepared under controlled conditions, may provide an enhanced protective effect against off-target doxorubicin toxicity.
Collapse
Affiliation(s)
- Sara Zaher
- Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut 71515, Egypt;
| | - Mahmoud E. Soliman
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt;
- Pharm D Program, Egypt-Japan University of Science and Technology (EJUST), New Borg El Arab, Alexandria 21934, Egypt
| | - Mahmoud Elsabahy
- School of Biotechnology and Science Academy, Badr University in Cairo, Badr City, Cairo 11829, Egypt;
| | - Rania M. Hathout
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt;
- Correspondence:
| |
Collapse
|
17
|
Ramkumar V, Sheth S, Dhukhwa A, Al Aameri R, Rybak L, Mukherjea D. Transient Receptor Potential Channels and Auditory Functions. Antioxid Redox Signal 2022; 36:1158-1170. [PMID: 34465184 PMCID: PMC9221156 DOI: 10.1089/ars.2021.0191] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Transient receptor potential (TRP) channels are cation-gated channels that serve as detectors of various sensory modalities, such as pain, heat, cold, and taste. These channels are expressed in the inner ear, suggesting that they could also contribute to the perception of sound. This review provides more details on the different types of TRP channels that have been identified in the cochlea to date, focusing on their cochlear distribution, regulation, and potential contributions to auditory functions. Recent Advances: To date, the effect of TRP channels on normal cochlear physiology in mammals is still unclear. These channels contribute, to a limited extent, to normal cochlear physiology such as the hair cell mechanoelectrical transduction channel and strial functions. More detailed information on a number of these channels in the cochlea awaits future studies. Several laboratories focusing on TRPV1 channels have shown that they are responsive to cochlear stressors, such as ototoxic drugs and noise, and regulate cytoprotective and/or cell death pathways. TRPV1 expression in the cochlea is under control of oxidative stress (produced primarily by NOX3 NADPH oxidase) as well as STAT1 and STAT3 transcription factors, which differentially modulate inflammatory and apoptotic signals in the cochlea. Inhibition of oxidative stress or inflammation reduces the expression of TRPV1 channels and protects against cochlear damage and hearing loss. Critical Issues: TRPV1 channels are activated by both capsaicin and cisplatin, which produce differential effects on the inner ear. How these differential actions are produced is yet to be determined. It is clear that TRPV1 is an essential component of cisplatin ototoxicity as knockdown of these channels protects against hearing loss. In contrast, activation of TRPV1 by capsaicin protected against subsequent hearing loss induced by cisplatin. The cellular targets that are influenced by these two drugs to account for their differential profiles need to be fully elucidated. Furthermore, the potential involvement of different TRP channels present in the cochlea in regulating cisplatin ototoxicity needs to be determined. Future Directions: TRPV1 has been shown to mediate the entry of aminoglycosides into the hair cells. Thus, novel otoprotective strategies could involve designing drugs to inhibit entry of aminoglycosides and possibly other ototoxins into cochlear hair cells. TRP channels, including TRPV1, are expressed on circulating and resident immune cells. These receptors modulate immune cell functions. However, whether they are activated by cochlear stressors to initiate cochlear inflammation and ototoxicity needs to be determined. A better understanding of the function and regulation of these TRP channels in the cochlea could enable development of novel treatments for treating hearing loss. Antioxid. Redox Signal. 36, 1158-1170.
Collapse
Affiliation(s)
- Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Sandeep Sheth
- Department of Pharmaceutical Sciences, Larkin University College of Pharmacy, Miami, Florida, USA
| | - Asmita Dhukhwa
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Raheem Al Aameri
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Leonard Rybak
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA.,Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Debashree Mukherjea
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| |
Collapse
|
18
|
Fetoni AR, Paciello F, Troiani D. Cisplatin Chemotherapy and Cochlear Damage: Otoprotective and Chemosensitization Properties of Polyphenols. Antioxid Redox Signal 2022; 36:1229-1245. [PMID: 34731023 DOI: 10.1089/ars.2021.0183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Significance: Cisplatin is an important component of treatment regimens for different cancers. Notwithstanding that therapeutic success often results from partial efficacy or stabilizing the disease, chemotherapy failure is driven by resistance to drug treatment and occurrence of side effects, such as progressive irreversible ototoxicity. Cisplatin's side effects, including ototoxicity, are often dose limiting. Recent Advances: Cisplatin ototoxicity results from several mechanisms, including redox imbalance caused by reactive oxygen species production and lipid peroxidation, activation of inflammation, and p53 and its downstream pathways that culminate in apoptosis. Considerable efforts in research have targeted development of molecular interventions that can be concurrently administered with cisplatin or other chemotherapies to reduce side effect toxicities while preserving or enhancing the antineoplastic effects. Evidence from studies has indicated some polyphenols, such as curcumin, can help to regulate redox signaling and inflammatory effects. Furthermore, polyphenols can exert opposing effects in different types of tissues, that is, normal cells undergoing stressful conditions versus cancer cells. Critical Issues: This review article summarizes evidence of curcumin antioxidant effect against cisplatin-induced ototoxicity that is converted to a pro-oxidant activity in cisplatin-treated cancer cells, thus providing an ideal chemosensitivity combined with otoprotection. Polyphenols can modulate the adaptive responses to stress in the cisplatin-exposed cochlea. These adaptive effects can result from the interaction/cross talk between the cell's defenses, inflammatory molecules, and the key signaling molecules of signal transducers and activators of transcription 3 (STAT-3), nuclear factor κ-B (NF-κB), p53, and nuclear factor erythroid 2-related factor 2 (Nrf-2). Future Directions: We provide molecular evidence for alternative strategies for chemotherapy with cisplatin addressing the otoprotection and chemosensitization properties of polyphenols. Antioxid. Redox Signal. 36, 1229-1245.
Collapse
Affiliation(s)
- Anna Rita Fetoni
- Department of Head and Neck Surgery, Università Cattolica Del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabiola Paciello
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diana Troiani
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
19
|
Amarasiri SS, Attanayake AP, Mudduwa LKB, Jayatilaka KAPW. Asparagus falcatus L. (Asparagaceae) leaf extracts attenuate doxorubicin-induced renal toxicity via antioxidant, anti-inflammatory, and anti-apoptotic pathways. Drug Chem Toxicol 2022:1-15. [PMID: 35637614 DOI: 10.1080/01480545.2022.2080218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The search for therapeutic agents that improve kidney function against doxorubicin-induced renal toxicity is important. Herein, the potential nephroprotective activity by Asparagus falcatus L. (AF, Asparagaceae) leaf extracts against doxorubicin-induced renal toxicity (5 mg/kg, ip) in Wistar rats (n = 6/group) after oral administration of hexane (55 mg/kg), ethyl acetate (35 mg/kg), butanol (75 mg/kg), and aqueous (200 mg/kg) extracts of AF for 28 consecutive days was investigated. It was noticed that the treatment with the selected extracts of AF significantly attenuated doxorubicin-induced elevations of serum creatinine, urea nitrogen, β2-microglobulin, cystatin C, and proteinuria in experimental rats. The histology showed attenuation of the features of acute tubular injury. Treatment regimens significantly reversed the doxorubicin-induced reduction in total antioxidant status, glutathione peroxidase, and glutathione reductase activity in renal tissue homogenates. A suppression in lipid peroxidation was noted with hexane, ethyl acetate, and butanol extracts of AF. Moreover, a reduction in the concentration of the pro-inflammatory mediator TNF-α (p < 0.05), and immunohistochemical expression of COX-2 were observed. The immunohistochemical expression of pro-apoptotic Bax protein was decreased and the anti-apoptotic BCL-2 was increased in renal tissues following the treatments. In conclusion, it was revealed that, hexane, ethyl acetate, butanol, and aqueous extracts of AF attenuate doxorubicin-induced renal toxicity in Wistar rats through antioxidant, anti-inflammatory, and anti-apoptotic pathways. The plant, AF could be recommended as a promising therapeutic agent to minimize renal toxicity induced by doxorubicin in cancer patients, however, subsequent clinical trials are warranted.
Collapse
Affiliation(s)
- Sachinthi S Amarasiri
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, University of Ruhuna, Galle, Sri Lanka
| | - Anoja P Attanayake
- Department of Biochemistry, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - Lakmini K B Mudduwa
- Department of Pathology, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | | |
Collapse
|
20
|
Shahcheraghi SH, Salemi F, Peirovi N, Ayatollahi J, Alam W, Khan H, Saso L. Nrf2 Regulation by Curcumin: Molecular Aspects for Therapeutic Prospects. Molecules 2021; 27:167. [PMID: 35011412 PMCID: PMC8746993 DOI: 10.3390/molecules27010167] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Nuclear factor erythroid 2 p45-related factor (2Nrf2) is an essential leucine zipper protein (bZIP) that is primarily located in the cytoplasm under physiological conditions. Nrf2 principally modulates endogenous defense in response to oxidative stress in the brain.In this regard, Nrf2 translocates into the nucleus and heterodimerizes with the tiny Maf or Jun proteins. It then attaches to certain DNA locations in the nucleus, such as electrophile response elements (EpRE) or antioxidant response elements (ARE), to start the transcription of cytoprotective genes. Many neoplasms have been shown to have over activated Nrf2, strongly suggesting that it is responsible for tumors with a poor prognosis. Exactly like curcumin, Zinc-curcumin Zn (II)-curc compound has been shown to induce Nrf2 activation. In the cancer cell lines analyzed, Zinc-curcumin Zn (II)-curc compound can also display anticancer effects via diverse molecular mechanisms, including markedly increasing heme oxygenase-1 (HO-1) p62/SQSTM1 and the Nrf2 protein levels along with its targets. It also strikingly decreases the levels of Nrf2 inhibitor, Kelch-like ECH-associated protein 1 (Keap1) protein.As a result, the crosstalk between p62/SQSTM1 and Nrf2 could be used to improve cancer patient response to treatments. The interconnected anti-inflammatory and antioxidative properties of curcumin resulted from its modulatory effects on Nrf2 signaling pathway have been shown to improve insulin resistance. Curcumin exerts its anti-inflammatory impact through suppressing metabolic reactions and proteins such as Keap1 that provoke inflammation and oxidation. A rational amount of curcumin-activated antioxidant Nrf2 HO-1 and Nrf2-Keap1 pathways and upregulated the modifier subunit of glutamate-cysteine ligase involved in the production of the intracellular antioxidant glutathione. Enhanced expression of glutamate-cysteine ligase, a modifier subunit (GLCM), inhibited transcription of glutamate-cysteine ligase, a catalytic subunit (GCLC). A variety of in vivo, in vitro and clinical studies has been done so far to confirm the protective role of curcumin via Nrf2 regulation. This manuscript is designed to provide a comprehensive review on the molecular aspects of curcumin and its derivatives/analogs via regulation of Nrf2 regulation.
Collapse
Affiliation(s)
- Seyed Hossein Shahcheraghi
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd 8916978477, Iran; (S.H.S.); (J.A.)
| | - Fateme Salemi
- School of Medicine, Islamic Azad University of Medical Sciences, Yazd 19395/1495, Iran;
| | - Niloufar Peirovi
- School of Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran;
| | - Jamshid Ayatollahi
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd 8916978477, Iran; (S.H.S.); (J.A.)
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, 00185 Rome, Italy;
| |
Collapse
|
21
|
Abarikwu SO, Njoku RCC, John IG, Amadi BA, Mgbudom-Okah CJ, Onuah CL. Antioxidant and anti-inflammatory protective effects of rutin and kolaviron against busulfan-induced testicular injuries in rats. Syst Biol Reprod Med 2021; 68:151-161. [PMID: 34753368 DOI: 10.1080/19396368.2021.1989727] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
There are few treatment options, including the use of natural phenolics-based combination therapy for mitigating male infertility conditions associated with chemotherapy. Busulfan is an anti-cancer drug that leads to testicular problems in humans. Here, we studied the effect of co-treatment of rutin and kolaviron against busulfan-induced testis damage. Young adult male Wistar rats were intraperitoneally injected busulfan (4 mg/kg b.w), and then orally administered rutin (30 mg/kg b.w), and kolaviron (50 mg/kg b.w) alone and combined for 60 days. Results revealed that rutin and kolaviron alone or in combination reversed busulfan-induced increase in oxidative stress along with sperm quality of treated animals. However, kolaviron and rutin separately improved the concentrations of MDA and GSH and sperm quality more than when they were combined. Similarly, rutin and kolaviron separately or in combination preserved spermatogenesis and relieved busulfan-induced increase in nitric oxide concentration, myeloperoxidase and 3β-hydroxysteroid dehydrogenase activities. Co-supplementation with kolaviron but not rutin nor when rutin was combined with kolaviron also improved the testicular level of tumor necrosis-alpha. Finally, the histological features in the testes caused by busulfan were reversed by rutin, whereas treatment with kolaviron alone or in combination with rutin partially protected the testis from busulfan-induced injury as demonstrated by the appearance of few germ cells, damaged tubules, loss of round spermatids and defoliation of the seminiferous epithelium. Thus, the combined treatment regimen of rutin and kolaviron sparingly prevented busulfan-induced testicular injuries in rats.Abbreviations: CAT: Catalase; GSH: Glutathione; 3β-HSD: 3β- hydroxysteroid Dehydrogenase; MDA: Malondialdehyde; TNF-α: Tumor necrosis-alpha; BUS: Busulfan; RUT: Rutin; KV: Kolaviron; TBARS: Thiobarbituric Acid Reactive Substances; MPO: Myeloperoxidase; ELISA: Enzyme-Linked Immunoassay; NAD: Nicotinamide Adenine Dinucleotide (oxidized); ROS: Reactive Oxygen Species.
Collapse
Affiliation(s)
- Sunny O Abarikwu
- Department of Biochemistry, University of Port Harcourt, Choba, Nigeria
| | - Rex-Clovis C Njoku
- Department of Chemistry/Biochemistry & Molecular Biology, Alex Ekwueme-Federal University Ndufu-Alike, Ikwo, Nigeria
| | - Ifeoma G John
- Department of Biochemistry, University of Port Harcourt, Choba, Nigeria
| | - Benjamin A Amadi
- Department of Biochemistry, University of Port Harcourt, Choba, Nigeria
| | | | - Chigozie L Onuah
- Department of Biochemistry, University of Port Harcourt, Choba, Nigeria
| |
Collapse
|
22
|
Alhilo I, Alhilo S, Alkhatib B, Al-Shorman A. Hyperthyroidism treatment by alternative therapies based on cupping and dietary-herbal supplementation: a case report. Drug Metab Pers Ther 2021; 37:95-98. [PMID: 35385890 DOI: 10.1515/dmpt-2021-0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/09/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Hyperthyroidism is characterized by increasing production of thyroid hormone (TH) and decreasing of thyroid stimulation hormone (TSH) secretion. The treatment of hyperthyroidism includes such as anti-thyroid drugs, radioiodine, and thyroidectomy have many side effects without complete curing results. We described a successful treatment of hyperthyroidism patient with dietary-herbal supplementation with wet cupping without any medicine. CASE PRESENTATION A 29-years female, blood analysis showed that she had low TSH (0.012 mlU/mL), and normal levels of T3 and T4. After completing 16 weeks on Carbimazole, TSH value still low (0.024 mlU/mL) and urticaria was appeared. She decided to stop Carbimazole and try alternative therapy choices. She received wet cupping and dietary-herbal supplementations (including royal jelly, green barley grass and Taraxaf®) for two months. Notably, TSH values was increased during-after intervention and urticaria was disappeared. CONCLUSIONS Alternative therapy could be a beneficial choice for hyperthyroidism treatment without any side effects or complications under physician supervision.
Collapse
Affiliation(s)
- Iman Alhilo
- Herbal Dynasty Medical Center, Mutah University, Amman, Jordan
| | - Samir Alhilo
- Herbal Dynasty Medical Center, The University of Jordan, Amman, Jordan
| | - Buthaina Alkhatib
- Department of Clinical Nutrition and Dietetics, Faculty of Science, Philadelphia University, Amman, Jordan
| | - Alaa Al-Shorman
- Human Nutrition and Dietetics, The University of Jordan, Amman, Jordan
| |
Collapse
|
23
|
Alhilo I, Alhilo S, Alkhatib B, Al-Shorman A. Hyperthyroidism treatment by alternative therapies based on cupping and dietary-herbal supplementation: a case report. Drug Metab Pers Ther 2021; 0:dmdi-2021-0132. [PMID: 34388334 DOI: 10.1515/dmdi-2021-0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/09/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Hyperthyroidism is characterized by increasing production of thyroid hormone (TH) and decreasing of thyroid stimulation hormone (TSH) secretion. The treatment of hyperthyroidism includes such as anti-thyroid drugs, radioiodine, and thyroidectomy have many side effects without complete curing results. We described a successful treatment of hyperthyroidism patient with dietary-herbal supplementation with wet cupping without any medicine. CASE PRESENTATION A 29-years female, blood analysis showed that she had low TSH (0.012 mlU/mL), and normal levels of T3 and T4. After completing 16 weeks on Carbimazole, TSH value still low (0.024 mlU/mL) and urticaria was appeared. She decided to stop Carbimazole and try alternative therapy choices. She received wet cupping and dietary-herbal supplementations (including royal jelly, green barley grass and Taraxaf®) for two months. Notably, TSH values was increased during-after intervention and urticaria was disappeared. CONCLUSIONS Alternative therapy could be a beneficial choice for hyperthyroidism treatment without any side effects or complications under physician supervision.
Collapse
Affiliation(s)
- Iman Alhilo
- Herbal Dynasty Medical Center, Mutah University, Amman, Jordan
| | - Samir Alhilo
- Herbal Dynasty Medical Center, The University of Jordan, Amman, Jordan
| | - Buthaina Alkhatib
- Department of Clinical Nutrition and Dietetics, Faculty of Science, Philadelphia University, Amman, Jordan
| | - Alaa Al-Shorman
- Human Nutrition and Dietetics, The University of Jordan, Amman, Jordan
| |
Collapse
|
24
|
Hussain Y, Islam L, Khan H, Filosa R, Aschner M, Javed S. Curcumin-cisplatin chemotherapy: A novel strategy in promoting chemotherapy efficacy and reducing side effects. Phytother Res 2021; 35:6514-6529. [PMID: 34347326 DOI: 10.1002/ptr.7225] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022]
Abstract
The efficacy of chemotherapy in cancer therapy is limited due to resistance, treatment selectivity, and severe adverse effects. Immunotherapy, chemotherapy, targeted therapy, radiation, and surgery are the most common therapeutic strategies for treatment, with chemotherapy being the most successful. Nonetheless, these treatments exhibit poor effectiveness due to toxicity and resistance. Therefore, combination therapies of natural products may be used as an effective and novel strategy to overcome such barriers. Cisplatin is a platinum-based chemotherapy agent, and when administered alone, it can lead to severe adverse effects and resistance mechanism resulting in therapeutic failure. Curcumin is a polyphenolic compound extracted from turmeric (Curcuma longa) exhibiting anticancer potential with minimal adverse effects. The combination therapy of curcumin and cisplatin is a novel strategy to mitigate/attenuate cisplatin-related adverse effects and improve the barrier of resistance reducing unwanted effects. However, there are uncertainties on the efficacy of curcumin, and more in depth and high-quality studies are needed. This review aims to explain the adverse effects related to individual cisplatin delivery, the positive outcome of individual curcumin delivery, and the combination therapy of curcumin and cisplatin from nano platform as a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Yaseen Hussain
- Lab of Controlled Release and Drug Delivery System, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Lubna Islam
- Department of Pharmacy, University of Malakand, Dir Lower Chakdara, KPK, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Rosanna Filosa
- Department of Experimental Medicine, University of Campania, "L. Vanvitelli", Naples, Italy
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Saba Javed
- Department of Zoology, Hazara University, Mansehra, Pakistan
| |
Collapse
|
25
|
Samra YA, Abdelghany AM, Zaghloul RA. Polydatin gold nanoparticles potentiate antitumor effect of doxorubicin in Ehrlich ascites carcinoma-bearing mice. J Biochem Mol Toxicol 2021; 35:e22869. [PMID: 34339076 DOI: 10.1002/jbt.22869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/16/2021] [Accepted: 07/19/2021] [Indexed: 12/19/2022]
Abstract
Breast cancer is a leading cause of death. Anticancer treatment such as gold nanoparticles (AuNP) seems highly promising in this regard. Therefore, this study aimed to assess the beneficial effect of doxorubicin (Dox) and polydatin (PD) AuNP in Ehrlich ascites carcinoma (EAC) and the ability of PD-AuNP to protect the heart from Dox's deteriorating effects. EAC was induced in mice. The mice were divided into nine groups: normal, EAC, PD: received PD (20 mg/kg), Dox: received Dox (2 mg/kg), PD-AuNPH: received 10 ppm AuNP of PD, PD-AuNPL: received 5 ppm AuNP of PD, Dox-AuNP: received Dox-AuNP, PD-Dox-AuNP: received PD-Dox-AuNP, AuNP: received AuNP. On the 21st day from tumor inoculation, the mice were sacrificed and tumor and heart tissues were removed. Tumor β-catenin/Cyclin D1 and p53 were assessed by immunohistochemistry. IL-6 was determined by enzyme-linked immunosorbent assay. PD-AuNP and Dox-AuNP showed a significant reduction in tumor volume and weight more than their free forms. Also, PD-AuNP and Dox-AuNP showed markedly less dense tumor cells. β-catenin and Cyclin D1 were markedly decreased and p53 was highly upregulated by PD-AuNP and Dox-AuNP. Moreover, PD-AuNP and Dox-AuNP have the ability to decrease IL-6 production. PD-AuNP protected the heart from Dox-induced severe degeneration. Therefore, PD-AuNP could be a tool to decelerate the progression of breast cancer.
Collapse
Affiliation(s)
- Yara A Samra
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Amr M Abdelghany
- Department of Spectroscopy, Physics Division, National Research Centre, Giza, Egypt.,Basic Science Department, Horus University, New Damietta, Damietta, Egypt
| | - Randa A Zaghloul
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
26
|
Wigner P, Bijak M, Saluk-Bijak J. The Green Anti-Cancer Weapon. The Role of Natural Compounds in Bladder Cancer Treatment. Int J Mol Sci 2021; 22:7787. [PMID: 34360552 PMCID: PMC8346071 DOI: 10.3390/ijms22157787] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/20/2022] Open
Abstract
Bladder cancer (BC) is the second most common genitourinary cancer. In 2018, 550,000 people in the world were diagnosed with BC, and the number of new cases continues to rise. BC is also characterized by high recurrence risk, despite therapies. Although in the last few years, the range of BC therapy has considerably widened, it is associated with severe side effects and the development of drug resistance, which is hampering treatment success. Thus, patients are increasingly choosing products of natural origin as an alternative or complementary therapeutic options. Therefore, in this article, we aim to elucidate, using the available literature, the role of natural substances such as curcumin, sulforaphane, resveratrol, quercetin, 6-gingerol, delphinidin, epigallocatechin-3-gallate and gossypol in the BC treatment. Numerous clinical and preclinical studies point to their role in the modulation of the signaling pathways, such as cell proliferation, cell survival, apoptosis and cell death.
Collapse
Affiliation(s)
- Paulina Wigner
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-136 Lodz, Poland;
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-136 Lodz, Poland;
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-136 Lodz, Poland;
| |
Collapse
|
27
|
Vora J, Athar M, Sinha S, Jha PC, Shrivastava N. Binding Insight of Anti-HIV Phytocompounds with Prime Targets of HIV: A Molecular Dynamics Simulation Analysis. Curr HIV Res 2021; 18:132-141. [PMID: 31995010 DOI: 10.2174/1570162x18666200129112509] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/27/2019] [Accepted: 01/09/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Despite intense efforts, AIDS is difficult to tackle by current anti-retroviral therapy (ART) due to its side effects; therefore, there is an urgent need to discover potential, multitarget and low-cost anti-HIV compounds. OBJECTIVE We have shown that few phytocompounds can potentially inhibit the prime targets of HIV namely GP120 envelope protein, reverse transcriptase, protease, integrase and ribonulcease. In this study, top ranked prioritized compounds were subjected to Molecular Dynamics (MD) simulation in order to study the conformational dynamics and integrity of crucial interaction in the receptor sites. METHODS The system was built for selected protein-ligand complex using TIP3P water model and OPLS_2005 force field. Trajectories were recorded up to 20 ns simulation time in Desmond module of Schrödinger software. RESULTS As a result of a comprehensive analysis of molecular properties and dynamics of the complexes, it has been concluded that Chebulic acid, Curcumin and Mulberroside C could be developed as envelope glycoprotein GP120 inhibitor, reverse transcriptase inhibitor and protease inhibitor respectively. However, the fluctuation of Chebulic acid with respect to integrase and ribonuclease protein was higher during the simulation. CONCLUSION These findings can aid in the designing of the structural properties for more effective anti-HIV compounds against the given targets.
Collapse
Affiliation(s)
- Jaykant Vora
- B.V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India.,Department of Life Science, Gujarat University, Ahmedabad, Gujarat; India
| | - Mohd Athar
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar, India
| | - Sonam Sinha
- B.V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India.,Department of Life Science, Gujarat University, Ahmedabad, Gujarat; India
| | - Prakash C Jha
- Centre for Applied Chemistry, Central University of Gujarat, Gandhinagar, India
| | - Neeta Shrivastava
- B.V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India
| |
Collapse
|
28
|
Bevacqua E, Curcio M, Saletta F, Vittorio O, Cirillo G, Tucci P. Dextran-Curcumin Nanosystems Inhibit Cell Growth and Migration Regulating the Epithelial to Mesenchymal Transition in Prostate Cancer Cells. Int J Mol Sci 2021; 22:7013. [PMID: 34209825 PMCID: PMC8269310 DOI: 10.3390/ijms22137013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/21/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
Functional nanocarriers which are able to simultaneously vectorize drugs to the site of interest and exert their own cytotoxic activity represent a significant breakthrough in the search for effective anticancer strategies with fewer side effects than conventional chemotherapeutics. Here, we propose previously developed, self-assembling dextran-curcumin nanoparticles for the treatment of prostate cancer in combination therapy with Doxorubicin (DOXO). Biological effectiveness was investigated by evaluating the cell viability in either cancer and normal cells, reactive oxygen species (ROS) production, apoptotic effect, interference with the cell cycle, and the ability to inhibit cell migration and reverse the epithelial to mesenchymal transition (EMT). The results proved a significant enhancement of curcumin efficiency upon immobilization in nanoparticles: IC50 reduced by a half, induction of apoptotic effect, and improved ROS production (from 67 to 134%) at low concentrations. Nanoparticles guaranteed a pH-dependent DOXO release, with a more efficient release in acidic environments. Finally, a synergistic effect between nanoparticles and Doxorubicin was demonstrated, with the free curcumin showing additive activity. Although in vivo studies are required to support the findings of this study, these preliminary in vitro data can be considered a proof of principle for the design of an effective therapy for prostate cancer treatment.
Collapse
Affiliation(s)
- Emilia Bevacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.B.); (M.C.); (G.C.)
| | - Manuela Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.B.); (M.C.); (G.C.)
| | - Federica Saletta
- Lowy Cancer Research Centre, Children’s Cancer Institute, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (F.S.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Orazio Vittorio
- Lowy Cancer Research Centre, Children’s Cancer Institute, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (F.S.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence for Convergent BioNano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.B.); (M.C.); (G.C.)
| | - Paola Tucci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (E.B.); (M.C.); (G.C.)
| |
Collapse
|
29
|
Li F, Liang Y, Wang M, Xu X, Zhao F, Wang X, Sun Y, Chen W. Multifunctional nanoplatforms as cascade-responsive drug-delivery carriers for effective synergistic chemo-photodynamic cancer treatment. J Nanobiotechnology 2021; 19:140. [PMID: 34001157 PMCID: PMC8130269 DOI: 10.1186/s12951-021-00876-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
Synergistic chemo-photodynamic therapy has garnered attention in the field of cancer treatment. Here, a pH cascade-responsive micellar nanoplatform with nucleus-targeted ability, for effective synergistic chemo-photodynamic cancer treatment, was fabricated. In this micellar nanoplatform, 5-(4-carboxyphenyl)-10,15,20-triphenylporphyrin (Por), a photodynamic therapy (PDT) agent was utilized for carrying the novel anticancer drug GNA002 to construct a hydrophobic core, and cyclic RGD peptide (cRGD)-modified polyethylene glycol (PEG) (cRGD-PEG) connected the cell-penetrating peptide hexaarginine (R6) through a pH-responsive hydrazone bond (cRGD-PEG-N = CH-R6) to serve as a hydrophilic shell for increasing blood circulation time. After passively accumulating in tumor sites, the self-assembled GNA002-loaded nanoparticles were actively internalized into cancer cells via the cRGD ligands. Once phagocytosed by lysosomes, the acidity-triggered detachment of the cRGD-PEG shell led to the formation of R6-coated secondary nanoparticles and subsequent R6-mediated nucleus-targeted drug delivery. Combined with GNA002-induced nucleus-specific chemotherapy, reactive oxygen species produced by Por under 532-nm laser irradiation achieved a potent synergistic chemo-photodynamic cancer treatment. Moreover, our in vitro and in vivo anticancer investigations revealed high cancer-suppression efficacy of this ideal multifunctional nanoplatform, indicating that it could be a promising candidate for synergistic anticancer therapy.![]()
Collapse
Affiliation(s)
- Fan Li
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Yan Liang
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China
| | - Miaochen Wang
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Xing Xu
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Fen Zhao
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China
| | - Xu Wang
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China.
| | - Yong Sun
- Department of Pharmaceutics, Qingdao University School of Pharmacy, Qingdao, 266021, China.
| | - Wantao Chen
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, China.
| |
Collapse
|
30
|
Identification of Beilschmiedia tsangii Root Extract as a Liver Cancer Cell-Normal Keratinocyte Dual-Selective NRF2 Regulator. Antioxidants (Basel) 2021; 10:antiox10040544. [PMID: 33915987 PMCID: PMC8066689 DOI: 10.3390/antiox10040544] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) plays a crucial role in regulating the expression of genes participating in cellular defense mechanisms against oxidative or xenobiotic insults. However, there is increasing evidence showing that hyperactivation of NRF2 is associated with chemoresistance in several cancers, including hepatocellular carcinoma (HCC), thus making NRF2 an attractive target for cancer therapy. Another important issue in cancer medication is the adverse effects of these substances on normal cells. Here, we attempted to identify a dual-selective NRF2 regulator that exerts opposite effects on NRF2-hyperactivated HCC cells and normal keratinocytes. An antioxidant response element driven luciferase reporter assay was established in Huh7 and HaCaT cells as high-throughput screening platforms. Screening of 3,000 crude extracts from the Taiwanese Indigenous Plant Extract Library resulted in the identification of Beilschmiedia tsangii (BT) root extract as a dual-selective NRF2 regulator. Multiple compounds were found to contribute to the dual-selective effects of BT extract on NRF2 signaling in two cell lines. BT extract reduced NRF2 protein level and target gene expression levels in Huh7 cells but increased them in HaCaT cells. Furthermore, notable combinatory cytotoxic effects of BT extract and sorafenib on Huh7 cells were observed. On the contrary, sorafenib-induced inflammatory reactions in HaCaT cells were reduced by BT extract. In conclusion, our results suggest that the combination of a selective NRF2 activator and inhibitor could be a practical strategy for fine-tuning NRF2 activity for better cancer treatment and that plant extracts or partially purified fractions could be a promising source for the discovery of dual-selective NRF2 regulators.
Collapse
|
31
|
Pandurangi RS, Tomasetti M, Verapazham ST, Paulmurugan R, Ma C, Rajput S, Anjanappa M, Nakshatri H. A Priori Activation of Apoptosis Pathways of Tumor (AAAPT) technology: Development of targeted apoptosis initiators for cancer treatment. PLoS One 2021; 16:e0225869. [PMID: 33556062 PMCID: PMC7870153 DOI: 10.1371/journal.pone.0225869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/18/2020] [Indexed: 11/24/2022] Open
Abstract
Cancer cells develop tactics to circumvent the interventions by desensitizing themselves to interventions. Amongst many, the principle routes of desensitization include a) activation of survival pathways (e.g. NF-kB, PARP) and b) downregulation of cell death pathways (e.g. CD95/CD95L). As a result, it requires high therapeutic dose to achieve tumor regression which, in turn damages normal cells through the collateral effects. Methods are needed to sensitize the low and non-responsive resistant tumor cells including cancer stem cells (CSCs) in order to evoke a better response from the current treatments. Current treatments including chemotherapy can induce cell death only in bulk cancer cells sparing CSCs and cancer resistant cells (CRCs) which are shown to be responsible for high recurrence of disease and low patient survival. Here, we report several novel tumor targeted sensitizers derived from the natural Vitamin E analogue (AMP-001-003). The drug design is based on a novel concept "A priori activation of apoptosis pathways of tumor technology (AAAPT) which is designed to activate specific cell death pathways and inhibit survival pathways simultaneously and selectively in cancer cells sparing normal cells. Our results indicate that AMP-001-003 sensitize various types of cancer cells including MDA-MB-231 (triple negative breast cancer), PC3 (prostate cancer) and A543 (lung cancer) cells resulting in reducing the IC-50 of doxorubicin in vitro when used as a combination. At higher doses, AMP-001 acts as an anti-tumor agent on its own. The synergy between AMP-001 and doxorubicin could pave a new pathway to use AAAPT leading molecules as neoadjuvant to chemotherapy to achieve better efficacy and reduced off-target toxicity compared to the current treatments.
Collapse
Affiliation(s)
- Raghu S. Pandurangi
- Sci-Engi-Medco Solutions Inc., St Charles, Missouri, United States of America
| | - Marco Tomasetti
- Department of Clinical and Molecular Sciences, Section of Experimental and Occupational Medicine, Polytechnic University of Marche, Ancona, Italy
| | - Sekar T. Verapazham
- Molecular Imaging Program at Stanford (MIPS), Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Cynthia Ma
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sandeep Rajput
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Manjushree Anjanappa
- Department Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Harikrishna Nakshatri
- Department Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
32
|
Famurewa AC, Edeogu CO, Offor FI, Besong EE, Akunna GG, Maduagwuna EK. Downregulation of redox imbalance and iNOS/NF-ĸB/caspase-3 signalling with zinc supplementation prevents urotoxicity of cyclophosphamide-induced hemorrhagic cystitis in rats. Life Sci 2020; 266:118913. [PMID: 33333050 DOI: 10.1016/j.lfs.2020.118913] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/23/2022]
Abstract
AIM Cyclophosphamide (CYP) chemotherapy induces bladder toxicity and hemorrhagic cystitis in cancer patients constituting a current clinical concern. Oxidative inflammatory cascades have been implicated as the mechanism contributing to CYP bladder urotoxicity. We thus assayed to explore whether zinc (Zn) supplementation could mitigate CYP-induced urotoxicity and evaluate the possible underlying mechanism in rats. MAIN METHOD Rats were orally administered Zn (100 mg/kg b.w./day) for 10 days against urotoxicity induced by single injection of CYP (150 mg/kg b.w., ip) on day 7. KEY FINDINGS CYP significantly depressed bladder activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and reduced glutathione (GSH) levels, whereas malondialdehyde level was increased prominently. In addition, CYP induced marked increases in the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and nitric oxide (NO) confirmed by histological alterations. CYP prominently increased bladder inducible nitric oxide synthase (iNOS) activity, nuclear factor-kappa B (NF-ĸB) and expression of caspase-3 protein. Zinc supplementation considerably abrogated the bladder urotoxicity by restoring redox balance, proinflammatory and apoptotic cascades and alleviated histopathological changes. SIGNIFICANCE This is the first to reveal zinc potential to prevent CYP-induced urotoxic hemorrhagic cystitis via restoring redox balance and enhancing anti-inflammatory and antiapoptotic mechanisms in rat bladder.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Ebonyi State, Nigeria.
| | - C O Edeogu
- Department of Medical Biochemistry, Faculty of Basic Medicine, Ebonyi State University, Abakaliki, Nigeria
| | - Florence I Offor
- Department of Medical Laboratory Sciences, Faculty of Health Sciences and Technology, Ebonyi State University, Abakaliki, Ebonyi State, Nigeria
| | - Elizabeth E Besong
- Department of Physiology, Faculty of Basic Medicine, Ebonyi State University, Abakaliki, Nigeria
| | - Gabriel G Akunna
- Department of Anatomy, College of Medicine and Health Sciences, Bowen University, Iwo, Osun State, Nigeria
| | | |
Collapse
|
33
|
Rahban M, Habibi-Rezaei M, Mazaheri M, Saso L, Moosavi-Movahedi AA. Anti-Viral Potential and Modulation of Nrf2 by Curcumin: Pharmacological Implications. Antioxidants (Basel) 2020; 9:E1228. [PMID: 33291560 PMCID: PMC7761780 DOI: 10.3390/antiox9121228] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is an essential transcription factor that maintains the cell's redox balance state and reduces inflammation in different adverse stresses. Under the oxidative stress, Nrf2 is separated from Kelch-like ECH-associated protein 1 (Keap1), which is a key sensor of oxidative stress, translocated to the nucleus, interacts with the antioxidant response element (ARE) in the target gene, and then activates the transcriptional pathway to ameliorate the cellular redox condition. Curcumin is a yellow polyphenolic curcuminoid from Curcuma longa (turmeric) that has revealed a broad spectrum of bioactivities, including antioxidant, anti-inflammatory, anti-tumor, and anti-viral activities. Curcumin significantly increases the nuclear expression levels and promotes the biological effects of Nrf2 via the interaction with Cys151 in Keap1, which makes it a marvelous therapeutic candidate against a broad range of oxidative stress-related diseases, including type 2 diabetes (T2D), neurodegenerative diseases (NDs), cardiovascular diseases (CVDs), cancers, viral infections, and more recently SARS-CoV-2. Currently, the multifactorial property of the diseases and lack of adequate medical treatment, especially in viral diseases, result in developing new strategies to finding potential drugs. Curcumin potentially opens up new views as possible Nrf2 activator. However, its low bioavailability that is due to low solubility and low stability in the physiological conditions is a significant challenge in the field of its efficient and effective utilization in medicinal purposes. In this review, we summarized recent studies on the potential effect of curcumin to activate Nrf2 as the design of potential drugs for a viral infection like SARS-Cov2 and acute and chronic inflammation diseases in order to improve the cells' protection.
Collapse
Affiliation(s)
- Mahdie Rahban
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran;
| | - Mehran Habibi-Rezaei
- School of Biology, College of Science, University of Tehran, Tehran 1417614335, Iran
- Center of Excellence in NanoBiomedicine, University of Tehran, Tehran 1417614335, Iran
| | - Mansoureh Mazaheri
- Research Center of Food Technology and Agricultural Products, Department of Food Toxicology, Standard Research Institute, Karaj 3158777871, Iran;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy;
| | - Ali A. Moosavi-Movahedi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran;
- UNESCO Chair on Interdisciplinary Research in Diabetes, University of Tehran, Tehran 1417614335, Iran
| |
Collapse
|
34
|
Visalli G, Facciolà A, Laganà P, Di Pietro A. Food chemoprevention and air pollution: the health comes with eating. REVIEWS ON ENVIRONMENTAL HEALTH 2020; 35:471-479. [PMID: 32573482 DOI: 10.1515/reveh-2019-0072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/13/2020] [Indexed: 06/11/2023]
Abstract
Ambient air pollution is known to be an important causative agent of many non-communicable diseases, mainly due to fine particulate matter (PM2.5). According to Global Burden Disease study in 2015, the estimated premature deaths caused by PM2.5 were 4.2 million. Besides deaths, airborne pollution's effect on human health also has dramatic economic and social costs, contributing greatly to disability-adjusted life-year (DALY). To reduce the health impact is necessary a double approach, which includes the improvement of air quality and food chemoprevention, aimed at enhancing the homeostatic abilities of exposed subjects. The scavenging, antioxidant, and anti-inflammatory properties of nutraceuticals effectively counteract the pathogenic mechanisms common in almost all non-communicable diseases associated with air pollutants. Moreover, several bioactive compounds of food modulate, by epigenetic mechanisms, the metabolism of xenobiotics, favouring conjugation reactions and promoting excretion. This narrative review summarize the numerous pieces of evidence collected in the last decades by observational and experimental studies which underline the chemopreventive role of flavonoids, contained in several fruits and consumer beverages (wine, tea, etc.), and isothiocyanate sulforaphane, contained in the cruciferous vegetables belonging to the genus Brassica. These bioactive compounds, enhancing the individual homeostatic abilities, reduce the harmful effects of airborne pollution.
Collapse
Affiliation(s)
- Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morpho Functional Imaging, University of Messina, Messina, Italy
| | - Alessio Facciolà
- Epidemiology Operative Unit, Department of Prevention, Health Provincial Agency, Messina, Italy
| | - Pasqualina Laganà
- Department of Biomedical and Dental Sciences and Morpho Functional Imaging, University of Messina, Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morpho Functional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
35
|
Polychemotherapy with Curcumin and Doxorubicin via Biological Nanoplatforms: Enhancing Antitumor Activity. Pharmaceutics 2020; 12:pharmaceutics12111084. [PMID: 33187385 PMCID: PMC7697177 DOI: 10.3390/pharmaceutics12111084] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/31/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOX) is a well-known chemotherapeutic agent extensively applied in the field of cancer therapy. However, similar to other chemotherapeutic agents such as cisplatin, paclitaxel, docetaxel, etoposide and oxaliplatin, cancer cells are able to obtain chemoresistance that limits DOX efficacy. In respect to dose-dependent side effect of DOX, enhancing its dosage is not recommended for effective cancer chemotherapy. Therefore, different strategies have been considered for reversing DOX resistance and diminishing its side effects. Phytochemical are potential candidates in this case due to their great pharmacological activities. Curcumin is a potential antitumor phytochemical isolated from Curcuma longa with capacity of suppressing cancer metastasis and proliferation and affecting molecular pathways. Experiments have demonstrated the potential of curcumin for inhibiting chemoresistance by downregulating oncogene pathways such as MMP-2, TGF-β, EMT, PI3K/Akt, NF-κB and AP-1. Furthermore, coadministration of curcumin and DOX potentiates apoptosis induction in cancer cells. In light of this, nanoplatforms have been employed for codelivery of curcumin and DOX. This results in promoting the bioavailability and internalization of the aforementioned active compounds in cancer cells and, consequently, enhancing their antitumor activity. Noteworthy, curcumin has been applied for reducing adverse effects of DOX on normal cells and tissues via reducing inflammation, oxidative stress and apoptosis. The current review highlights the anticancer mechanism, side effects and codelivery of curcumin and DOX via nanovehicles.
Collapse
|
36
|
Famurewa AC, Ekeleme-Egedigwe CA, Onwe CS, Egedigwe UO, Okoro CO, Egedigwe UJ, Asogwa NT. Ginger juice prevents cisplatin-induced oxidative stress, endocrine imbalance and NO/iNOS/NF-κB signalling via modulating testicular redox-inflammatory mechanism in rats. Andrologia 2020; 52:e13786. [PMID: 32777091 DOI: 10.1111/and.13786] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/11/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
The off-target testicular toxicity of the anticancer drug, cisplatin, is a current clinical concern and worrisome to male cancer patients. Growing evidence has implicated oxidative stress and inflammation in cisplatin toxicity. We have explored whether fresh ginger juice could mitigate testicular toxicity induced by anticancer drug cisplatin in rats. Rats were subjected to oral administration of fresh ginger juice (5 ml/kg body weight/day) for 5 days against testicular damage induced by single ip injection of cisplatin (CIS) (10 mg/kg body weight) on day 2 only. Testicular activities of antioxidant enzymes, malondialdehyde (MDA), reduced glutathione (GSH), nitric oxide (NO), inflammatory cytokines, inducible nitric oxide synthase (iNOS) and nuclear factor-ĸB (NF-ĸB) and serum hormone levels were estimated. CIS-induced prominent decreases in antioxidant enzyme activities, GSH and serum hormone levels, whereas levels of MDA, cytokines, NO, iNOS and NF-ĸB increased remarkably (p < .05) compared to control. Interestingly, the CIS-induced testicular alterations were considerably mitigated by the fresh ginger juice via abrogation of oxidative stress and anti-inflammatory mechanism. The study suggests, for the first time, antioxidant and anti-inflammatory effects of ginger juice against CIS testicular damage. Fresh ginger juice may have beneficial health impact on testicular side effect of CIS chemotherapy.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Chima A Ekeleme-Egedigwe
- Department of Chemistry/Biochemistry and Molecular Biology, Faculty of Science, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Chikodili S Onwe
- Department of Applied Biology, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | - Uchenna O Egedigwe
- Department of Plant Science and Biotechnology, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
| | - Chukwuemeka O Okoro
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Akakaliki, Nigeria
| | - Ugochukwu J Egedigwe
- Department of Haematology, University of Abuja Teaching Hospital, Abuja, Nigeria
| | | |
Collapse
|
37
|
Yu X, Yu W, Han X, Chen Z, Wang S, Zhai H. Sensitive analysis of doxorubicin and curcumin by micellar electromagnetic chromatography with a double wavelength excitation source. Anal Bioanal Chem 2020; 413:469-478. [PMID: 33118040 DOI: 10.1007/s00216-020-03017-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/06/2020] [Accepted: 10/16/2020] [Indexed: 12/01/2022]
Abstract
Doxorubicin has been extensively used to treat cancers, and there are recent findings that the anticancer activities can be enhanced by curcumin. Although the two compounds have native fluorescence, they can hardly be quantified directly simultaneously using the laser-induced fluorescence (LIF) detection method. To avoid complex fluorescence derivatization and introduction of interfering components, a highly sensitive double wavelength excitation source LIF (D-W-Ex-LIF) detector composed of a 445-nm and 488-nm commercial laser diode was constructed to detect them simultaneously. Rhodamine 6G was selected as an internal standard, because its fluorescence can be excited at 445 nm and 488 nm. The native fluorescence of doxorubicin and curcumin and their resolution were enhanced by introducing mixed micelles. The optimal electrophoretic separation buffer was 10 mM borate buffer containing 20 mM Triton X-100, 5 mM sodium dodecyl sulfate, and 30% (v/v) methanol at pH 9.00. Therefore, the developed method was specific, accurate, and easily operable. Its limits of detection for doxorubicin and curcumin in human urine samples were 4.00 × 10-3 and 1.00 × 10-2 μg/mL, respectively, and the limits of quantification were 1.00 × 10-2 and 3.00 × 10-2 μg/mL, respectively. The recoveries were 94.9-109.1%. Graphical abstract.
Collapse
Affiliation(s)
- Xiao Yu
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Wanxiang Yu
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiufen Han
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zuanguang Chen
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shumei Wang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Haiyun Zhai
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
38
|
Zhang P, Li T, Liu C, Sindi M, Cheng X, Qi S, Liu X, Yan Y, Bao Y, Brand-Saberi B, Yang W, Wang G, Yang X. Nano-sulforaphane attenuates PhIP-induced early abnormal embryonic neuro-development. Ann Anat 2020; 233:151617. [PMID: 33098981 DOI: 10.1016/j.aanat.2020.151617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND 2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyrimidine (PhIP), one of the most abundant heterocyclic aromatic amines (HAA) formed by cooking meat at high temperatures, may modify humans and rodents through the metabolic process prior to affecting nervous system development. In humans and rodents may be modified by metabolic processes and then affecting nervous system development. METHODS In this paper, PhIP was used to prepare a chicken embryo model with abnormal embryonic nervous system defects. Sulforaphane (SFN) is a derivative of a glucosinolate, which is abundant in cruciferous vegetables, and can pass through the placental barrier. Moreover, SFN has antioxidant and anti-apoptotic functions and is considered as a bioactive antioxidant with significant neuroprotective effects. Nano-sulforaphane (Nano-SFN, sulforaphane nanoparticles) was prepared by self-assembly using biocompatible, biodegradable methoxy polyethylene glycol 5000-b-polyglutamic acid 10,000 (mPEG5K-PGA10K) as the substrate, to explore the new application of Nano-SFN and its modified compounds as leading compounds in protecting against the abnormal development of the embryonic nervous system. RESULTS The results show that Nano-SFN could protect against PhIP-induced central nervous system (CNS, derived from neural tube) and peripheral nervous system (PNS, derived from neural crest cells, NCCs) defects and neural tube defects (NTDs), and increase the embryo survival rate. CONCLUSIONS This study indicates that Nano-SFN can effectively alleviate the developmental defects of embryonic nervous system induced by PhIP in the microenvironment and has a protective effect on embryonic development. It not only helps with expanding the application of SFN and improving its medicinal value, but also provides a possibility of SFN being developed as a novel drug for neuroprotection.
Collapse
Affiliation(s)
- Ping Zhang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Tingting Li
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Chang Liu
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Mustafa Sindi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Xin Cheng
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Shuangyu Qi
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Xinyue Liu
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Yu Yan
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7UQ, UK
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Weidong Yang
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China.
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
39
|
Yeung PK, Mohammadizadeh S, Akhoundi F, Mann K, Agu RU, Pulinilkunnil T. Hemodynamic Assessment and in vivo Catabolism of Adenosine 5'- Triphosphate in Doxorubicin or Isoproterenol-induced Cardiovascular Toxicity. Drug Metab Lett 2020; 14:80-88. [PMID: 33092518 DOI: 10.2174/1872312814666201022103802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/18/2020] [Accepted: 08/30/2020] [Indexed: 11/22/2022]
Abstract
PURPOSE Previous studies have shown catabolism of adenosine 5'-triphosphate (ATP) in systemic blood is a potential surrogate biomarker for cardiovascular toxicity. We compared the acute toxicity of high doses of doxorubicin (DOX) and isoproterenol (ISO) on hemodynamics and ATP catabolism in systemic circulation. METHODS Sprague Dawley (SD) rats (n = 8 - 11) were each given either a single dose of 30 mg/kg ISO, or twice-daily dose of 10 mg/kg of DOX or normal saline (control) for 4 doses by subcutaneous injection. Blood samples were collected up to 6 hours for measuring concentrations of ATP and its catabolites. Hemodynmics was recorded continuously. Difference was considered significant at p < 0.05 (ANOVA). RESULTS AND DISCUSSION Mortality was 1/8, 5/11 and 0/11 for the DOX, ISO and control groups, respectively. Systolic blood pressure was significantly lower in the DOX and ISO treated rats than in the control measured at the last recorded time (76 ± 9 for DOX vs 42 ± 8 for ISO vs 103 ± 5 mmHg for Control, p < 0.05 for all). Blood pressure fell gradually after the final injection for both DOX and control groups, but abruptly after ISO followed by a rebound and then gradual decline till the end of the experiment. Heart rate was significantly higher after ISO, but no difference between the DOX and control rats (p > 0.05). RBC concentrations of ADP and AMP, and plasma concentrations of adenosine and uric acid were significantly higher in the ISO group. In contrast, hypoxanthine concentrations were significantly higher in the DOX treated group (p < 0.05). CONCLUSION Acute cardiovascular toxicity induced by DOX and ISO may be measured by changes in hemodynamics and breakdown of ATP and adenosine in the systemic circulation, albeit a notable qualitative and quantitative difference was observed.
Collapse
Affiliation(s)
- Pollen K Yeung
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Faculties of Health and Medicine, Dalhousie University, Halifax, NS. Canada
| | - Sheyda Mohammadizadeh
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Faculties of Health and Medicine, Dalhousie University, Halifax, NS. Canada
| | - Fatemeh Akhoundi
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Faculties of Health and Medicine, Dalhousie University, Halifax, NS. Canada
| | - Kelsey Mann
- Pharmacokinetics and Metabolism Laboratory, College of Pharmacy and Department of Medicine, Faculties of Health and Medicine, Dalhousie University, Halifax, NS. Canada
| | - Remigius U Agu
- Biopharmaceutics and Drug Delivery Laboratory, College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, NS. Canada
| | - Thomas Pulinilkunnil
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB. Canada
| |
Collapse
|
40
|
Zhou X, Zijlstra SN, Soto-Gamez A, Setroikromo R, Quax WJ. Artemisinin Derivatives Stimulate DR5-Specific TRAIL-Induced Apoptosis by Regulating Wildtype P53. Cancers (Basel) 2020; 12:E2514. [PMID: 32899699 PMCID: PMC7563660 DOI: 10.3390/cancers12092514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 12/26/2022] Open
Abstract
Artemisinin derivatives, widely known as commercial anti-malaria drugs, may also have huge potential in treating cancer cells. It has been reported that artemisinin derivatives can overcome resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in liver and cervical cancer cells. In our study, we demonstrated that artesunate (ATS) and dihydroartemisinin (DHA) are more efficient in killing colon cancer cells compared to artemisinin (ART). ATS/DHA induces the expression of DR5 in a P53 dependent manner in HCT116 and DLD-1 cells. Both ATS and DHA overcome the resistance to DHER-induced apoptosis in HCT116, mainly through upregulating death receptor 5 (DR5). We also demonstrate that DHA sensitizes HCT116 cells to DHER-induced apoptosis via P53 regulated DR5 expression in P53 knockdown assays. Nevertheless, a lower effect was observed in DLD-1 cells, which has a single Ser241Phe mutation in the P53 DNA binding domain. Thus, the status of P53 could be one of the determinants of TRAIL resistance in some cancer cells. Finally, the combination treatment of DHA and the TRAIL variant DHER increases cell death in 3D colon cancer spheroid models, which shows its potential as a novel therapy.
Collapse
Affiliation(s)
| | | | | | | | - Wim J. Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (S.N.Z.); (A.S.-G.); (R.S.)
| |
Collapse
|
41
|
Abstract
Covering: up to 2020The transcription factor NRF2 is one of the body's major defense mechanisms, driving transcription of >300 antioxidant response element (ARE)-regulated genes that are involved in many critical cellular processes including redox regulation, proteostasis, xenobiotic detoxification, and primary metabolism. The transcription factor NRF2 and natural products have an intimately entwined history, as the discovery of NRF2 and much of its rich biology were revealed using natural products both intentionally and unintentionally. In addition, in the last decade a more sinister aspect of NRF2 biology has been revealed. NRF2 is normally present at very low cellular levels and only activated when needed, however, it has been recently revealed that chronic, high levels of NRF2 can lead to diseases such as diabetes and cancer, and may play a role in other diseases. Again, this "dark side" of NRF2 was revealed and studied largely using a natural product, the quassinoid, brusatol. In the present review, we provide an overview of NRF2 structure and function to orient the general reader, we will discuss the history of NRF2 and NRF2-activating compounds and the biology these have revealed, and we will delve into the dark side of NRF2 and contemporary issues related to the dark side biology and the role of natural products in dissecting this biology.
Collapse
Affiliation(s)
- Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA.
| | | |
Collapse
|
42
|
Rutz J, Janicova A, Woidacki K, Chun FKH, Blaheta RA, Relja B. Curcumin-A Viable Agent for Better Bladder Cancer Treatment. Int J Mol Sci 2020; 21:ijms21113761. [PMID: 32466578 PMCID: PMC7312715 DOI: 10.3390/ijms21113761] [Citation(s) in RCA: 20] [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: 04/22/2020] [Revised: 05/12/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Although the therapeutic armamentarium for bladder cancer has considerably widened in the last few years, severe side effects and the development of resistance hamper long-term treatment success. Thus, patients turn to natural plant products as alternative or complementary therapeutic options. One of these is curcumin, the principal component of Curcuma longa that has shown chemopreventive effects in experimental cancer models. Clinical and preclinical studies point to its role as a chemosensitizer, and it has been shown to protect organs from toxicity induced by chemotherapy. These properties indicate that curcumin could hold promise as a candidate for additive cancer treatment. This review evaluates the relevance of curcumin as an integral part of therapy for bladder cancer.
Collapse
Affiliation(s)
- Jochen Rutz
- Department of Urology, Goethe-University, 60438 Frankfurt am Main, Germany; (J.R.); (F.K.-H.C.)
| | - Andrea Janicova
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, 39106 Magdeburg, Germany; (A.J.); (K.W.); (B.R.)
| | - Katja Woidacki
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, 39106 Magdeburg, Germany; (A.J.); (K.W.); (B.R.)
| | - Felix K.-H. Chun
- Department of Urology, Goethe-University, 60438 Frankfurt am Main, Germany; (J.R.); (F.K.-H.C.)
| | - Roman A. Blaheta
- Department of Urology, Goethe-University, 60438 Frankfurt am Main, Germany; (J.R.); (F.K.-H.C.)
- Correspondence:
| | - Borna Relja
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, 39106 Magdeburg, Germany; (A.J.); (K.W.); (B.R.)
| |
Collapse
|
43
|
Chen Y, Deng Y, Zhu C, Xiang C. Anti prostate cancer therapy: Aptamer-functionalized, curcumin and cabazitaxel co-delivered, tumor targeted lipid-polymer hybrid nanoparticles. Biomed Pharmacother 2020; 127:110181. [PMID: 32416561 DOI: 10.1016/j.biopha.2020.110181] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/12/2020] [Accepted: 04/17/2020] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PC) is the most common type of newly diagnosed malignancy in men. Combined chemotherapy has been shown to be an effective strategy for the treatment of PC therapy. Lipid-polymer hybrid nanoparticles (LPNs) are core-shell nanoparticles composed of a polymer core and a lipid shell, which are reported to provide significant advantages for combined PC therapy. This study synthesized an aptamer conjugated ligand and designed an aptamer-functionalized, curcumin (CUR) and cabazitaxel (CTX) co-delivered LPNs (APT-CUR/CTX-LPNs). APT-CUR/CTX-LPNs had a mean size of 121.3 ± 4.2 nm and a positive surface charge (23.5 ± 2.6 mV). Both CUR and CTX were sustained released from LPNs. Aptamer-functionalized APT-CUR/CTX-LPNs exhibited good cell inhibition ability, high tumor accumulation, and remarkable tumor inhibition efficiency at the drug ratio of 2:5 (CUR:CTX). The novel LPNs offers great promise for the double drugs delivery to the prostate cancer cells and tumor xenograft in vivo, showing the potential of synergistic combination therapy for prostate cancer.
Collapse
Affiliation(s)
- Yougan Chen
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi 214062, PR China
| | - Yuanyuan Deng
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi 214062, PR China
| | - Chenyao Zhu
- Shenzhen Yuce Biotechnology Co. Ltd, Shenzhen 518000, PR China
| | - Congming Xiang
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi 214062, PR China.
| |
Collapse
|
44
|
Calcabrini C, Maffei F, Turrini E, Fimognari C. Sulforaphane Potentiates Anticancer Effects of Doxorubicin and Cisplatin and Mitigates Their Toxic Effects. Front Pharmacol 2020; 11:567. [PMID: 32425794 PMCID: PMC7207042 DOI: 10.3389/fphar.2020.00567] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
The success of cancer therapy is often compromised by the narrow therapeutic index of many anticancer drugs and the occurrence of drug resistance. The association of anticancer therapies with natural compounds is an emerging strategy to improve the pharmaco-toxicological profile of cancer chemotherapy. Sulforaphane, a phytochemical found in cruciferous vegetables, targets multiple pathways involved in cancer development, as recorded in different cancers such as breast, brain, blood, colon, lung, prostate, and so forth. As examples to make the potentialities of the association chemotherapy raise, here we highlight and critically analyze the information available for two associations, each composed by a paradigmatic anticancer drug (cisplatin or doxorubicin) and sulforaphane.
Collapse
Affiliation(s)
- Cinzia Calcabrini
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Francesca Maffei
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Eleonora Turrini
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| |
Collapse
|
45
|
Potential Applications of NRF2 Modulators in Cancer Therapy. Antioxidants (Basel) 2020; 9:antiox9030193. [PMID: 32106613 PMCID: PMC7139512 DOI: 10.3390/antiox9030193] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/17/2023] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (NRF2)-Kelch-like ECH-associated protein 1 (KEAP1) regulatory pathway plays an essential role in protecting cells and tissues from oxidative, electrophilic, and xenobiotic stress. By controlling the transactivation of over 500 cytoprotective genes, the NRF2 transcription factor has been implicated in the physiopathology of several human diseases, including cancer. In this respect, accumulating evidence indicates that NRF2 can act as a double-edged sword, being able to mediate tumor suppressive or pro-oncogenic functions, depending on the specific biological context of its activation. Thus, a better understanding of the mechanisms that control NRF2 functions and the most appropriate context of its activation is a prerequisite for the development of effective therapeutic strategies based on NRF2 modulation. In line of principle, the controlled activation of NRF2 might reduce the risk of cancer initiation and development in normal cells by scavenging reactive-oxygen species (ROS) and by preventing genomic instability through decreased DNA damage. In contrast however, already transformed cells with constitutive or prolonged activation of NRF2 signaling might represent a major clinical hurdle and exhibit an aggressive phenotype characterized by therapy resistance and unfavorable prognosis, requiring the use of NRF2 inhibitors. In this review, we will focus on the dual roles of the NRF2-KEAP1 pathway in cancer promotion and inhibition, describing the mechanisms of its activation and potential therapeutic strategies based on the use of context-specific modulation of NRF2.
Collapse
|
46
|
Rafiei H, Ashrafizadeh M, Ahmadi Z. MicroRNAs as novel targets of sulforaphane in cancer therapy: The beginning of a new tale? Phytother Res 2020; 34:721-728. [PMID: 31972874 DOI: 10.1002/ptr.6572] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/30/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
Abstract
Effective management and treatment of cancer depend on developing novel antitumor drugs with the capability of targeting various molecular pathways. Identification and subsequent targeting of these pathways are of importance in cancer therapy. MicroRNAs (miRNAs) are small noncoding RNA molecules responsible for post-transcriptional regulation of genes. Notably, miRNAs participate in a number of biological processes such as proliferation, apoptosis, differentiation, and cell cycle regulation. So, any impairment in the expression and function of miRNAs is associated with development of disorders, particularly cancer. Naturally occurring nutraceutical compounds have attracted much attention due to their great antitumor activity. Among them, sulforaphane isolated from Brassica oleracea (broccoli) is of interest due to its therapeutic and biological activities such as antidiabetic, antioxidant, anti-inflammatory, hepatoprotection, and cardiprotection. Sulforaphane has demonstrated great antitumor activity and is able to significantly inhibit proliferation, viability, migration, malignancy, and epithelial-to-mesenchymal transition of cancer cells. These antitumor effects have widely been investigated, and it appears that there is a need for a precise review to demonstrate the molecular pathway that sulforaphane follows to exert its antitumor activity. At the present review, we focus on the modulatory impact of sulforaphane on miRNAs and exhibit that how various miRNAs in different cancers are regulated by sulforaphane.
Collapse
Affiliation(s)
- Hossein Rafiei
- Department of Biology, Faculty of Sciences, Islamic Azad University, Shiraz Branch, Shiraz, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Zahra Ahmadi
- Department of basic science, Islamic Azad university, Shoushtar Branch, Shoushtar, Iran
| |
Collapse
|
47
|
Curcumin and Its Derivatives as Potential Therapeutic Agents in Prostate, Colon and Breast Cancers. Molecules 2019; 24:molecules24234386. [PMID: 31801262 PMCID: PMC6930580 DOI: 10.3390/molecules24234386] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/27/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer is a life-threatening disease and is the second leading cause of death around the world. The increasing threats of drug-resistant cancers indicate that there is an urgent need for the improvement or development of more effective anticancer agents. Curcumin, a phenolic compound originally derived from turmeric plant (Curcuma longa L. (Zingiberaceae family)) widely known as a spice and a coloring agent for food have been reported to possess notable anticancer activity by inhibiting the proliferation and metastasis, and enhancing cell cycle arrest or apoptosis in various cancer cells. In spite of all these benefits, the therapeutic application of curcumin in clinical medicine and its bioavailability are still limited due to its poor absorption and rapid metabolism. Structural modification of curcumin through the synthesis of curcumin-based derivatives is a potential approach to overcome the above limitations. Curcumin derivatives can overcome the disadvantages of curcumin while enhancing the overall efficacy and hindering drug resistance. This article reports a review of published curcumin derivatives and their enhanced anticancer activities.
Collapse
|
48
|
Ferreira SBDS, Braga G, Oliveira ÉL, da Silva JB, Rosseto HC, de Castro Hoshino LV, Baesso ML, Caetano W, Murdoch C, Colley HE, Bruschi ML. Design of a nanostructured mucoadhesive system containing curcumin for buccal application: from physicochemical to biological aspects. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2019; 10:2304-2328. [PMID: 31886108 PMCID: PMC6902884 DOI: 10.3762/bjnano.10.222] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/23/2019] [Indexed: 05/04/2023]
Abstract
Mucoadhesive nanostructured systems comprising poloxamer 407 and Carbopol 974P® have already demonstrated good mucoadhesion, as well as improved mechanical and rheological properties. Curcumin displays excellent biological activity, mainly in oral squamous cancer; however, its physicochemical characteristics hinder its application. Therefore, the aim of this study was to develop nanostructured formulations containing curcumin for oral cancer therapy. The photophysical interactions between curcumin and the formulations were elucidated by incorporation kinetics and location studies. They revealed that the drug was quickly incorporated and located in the hydrophobic portion of nanometer-sized polymeric micelles. Moreover, the systems displayed plastic behavior with rheopexy characteristics at 37 °C, viscoelastic properties and a gelation temperature of 36 °C, which ensures increased retention after application in the oral cavity. The mucoadhesion results confirmed the previous findings with the nanostructured systems showing a residence time of 20 min in porcine oral mucosa under flow system conditions. Curcumin was released after 8 h and could permeate through the porcine oral mucosa. Cytotoxicity testing revealed that the formulations were selective to cancer cells over healthy cells. Therefore, these systems could improve the physicochemical characteristics of curcumin by providing improved release and permeation, while selectivity targeting cancer cells.
Collapse
Affiliation(s)
- Sabrina Barbosa de Souza Ferreira
- Laboratory of Research and Development of Drug Delivery Systems, Postgraduate Program in Pharmaceutical Sciences, Department of Pharmacy, State University of Maringa, Maringa, Brazil
| | - Gustavo Braga
- Department of Chemistry, State University of Maringa, Maringa, Brazil
| | | | - Jéssica Bassi da Silva
- Laboratory of Research and Development of Drug Delivery Systems, Postgraduate Program in Pharmaceutical Sciences, Department of Pharmacy, State University of Maringa, Maringa, Brazil
| | - Hélen Cássia Rosseto
- Laboratory of Research and Development of Drug Delivery Systems, Postgraduate Program in Pharmaceutical Sciences, Department of Pharmacy, State University of Maringa, Maringa, Brazil
| | | | | | - Wilker Caetano
- Department of Chemistry, State University of Maringa, Maringa, Brazil
| | - Craig Murdoch
- The School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| | | | - Marcos Luciano Bruschi
- Laboratory of Research and Development of Drug Delivery Systems, Postgraduate Program in Pharmaceutical Sciences, Department of Pharmacy, State University of Maringa, Maringa, Brazil
| |
Collapse
|