1
|
Maji A, Paul A, Sarkar A, Nahar S, Bhowmik R, Samanta A, Nahata P, Ghosh B, Karmakar S, Kumar Maity T. Significance of TRAIL/Apo-2 ligand and its death receptors in apoptosis and necroptosis signalling: Implications for cancer-targeted therapeutics. Biochem Pharmacol 2024; 221:116041. [PMID: 38316367 DOI: 10.1016/j.bcp.2024.116041] [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: 10/03/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
The human immune defensesystem routinely expresses the tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), which is the most prevalent element for antitumor immunity. TRAIL associates with its death receptors (DRs), DR4 (TRAIL-R1), and DR5 (TRAIL-R2), in cancer cells to initiate the intracellular apoptosis cascade. Accordingly, numerous academic institutions and pharmaceutical companies havetried to exploreTRAIL's capacity to kill tumourcells by producing recombinant versions of it (rhTRAIL) or TRAIL receptor agonists (TRAs) [monoclonal antibody (mAb), synthetic and natural compounds, etc.] and molecules that sensitize TRAIL signalling pathway for therapeutic applications. Recently, several microRNAs (miRs) have been found to activate or inhibit death receptor signalling. Therefore, pharmacological regulation of these miRs may activate or resensitize the TRAIL DRs signal, and this is a novel approach for developing anticancer therapeutics. In this article, we will discuss TRAIL and its receptors and molecular pathways by which it induces various cell death events. We will unravel potential innovative applications of TRAIL-based therapeutics, and other investigated therapeutics targeting TRAIL-DRs and summarize the current preclinical pharmacological studies and clinical trials. Moreover, we will also emphasizea few situations where future efforts may be addressed to modulate the TRAIL signalling pathway.
Collapse
Affiliation(s)
- Avik Maji
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Abhik Paul
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Arnab Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India; Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata-700032, India.
| | - Sourin Nahar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Rudranil Bhowmik
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India; Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata-700032, India.
| | - Ajeya Samanta
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Pankaj Nahata
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad-500078, India.
| | - Sanmoy Karmakar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India; Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata-700032, India.
| | - Tapan Kumar Maity
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| |
Collapse
|
2
|
Alsina-Sánchez Á, Montalvo-Vázquez S, Grafals-Ruiz N, Acosta C, Ormé EM, Rodríguez I, Delgado-Rivera SM, Tinoco AD, Dharmawardhane S, Montes-González IC. Synthesis of Novel Heterocyclic Ferrocenyl Chalcones and Their Biological Evaluation. ACS OMEGA 2023; 8:34377-34387. [PMID: 37779926 PMCID: PMC10534901 DOI: 10.1021/acsomega.3c01830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/17/2023] [Indexed: 10/03/2023]
Abstract
Breast cancer is currently the most commonly diagnosed cancer, with 287,850 new cases estimated for 2022 as reported by the American Cancer Society. Therefore, finding an effective treatment for this disease is imperative. Chalcones are α,β-unsaturated systems found in nature. These compounds have shown a wide array of biological activities, making them popular synthetic targets. Chalcones consist of two aromatic substituents connected by an enone bridge; this arrangement allows for a large number of derivatives. Given the biological relevance of these compounds, novel ferrocene-heterocycle-containing chalcones were synthesized and characterized based on a hybrid drug design approach. These heterocycles included thiophene, pyrimidine, thiazolyl, and indole groups. Fourteen novel heterocyclic ferrocenyl chalcones were synthesized and characterized. Herein, we also report their cytotoxicity against triple-negative breast cancer cell lines MDA-MB-231 and 4T1 and the noncancer lung cell line MRC-5. System 3 ferrocenyl chalcones displayed superior anticancer properties compared to their system 1 analogues. System 3 chalcones bearing five-membered heterocyclic substituents (thiophene, pyrazole, pyrrole, and pyrimidine) were the most active toward the MDA-MB-231 cancer cell line with IC50 values from 6.59 to 12.51 μM. Cytotoxicity of the evaluated compounds in the 4T1 cell line exhibited IC50 values from 13.23 to 213.7 μM. System 3 pyrazole chalcone had consistent toxicity toward both cell lines (IC50 ∼ 13 μM) as well as promising selectivity relative to the noncancer MRC-5 control. Antioxidant activity was also evaluated, where, contrary to anticancer capabilities, system 1 ferrocenyl chalcones were superior to their system 3 analogues. Antioxidant activity comparable to that of ascorbic acid was observed for thiophene-bearing ferrocenyl chalcone with EC50 = 31 μM.
Collapse
Affiliation(s)
- Ámbar
M. Alsina-Sánchez
- Department
of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan 00931, Puerto Rico
| | | | - Nilmary Grafals-Ruiz
- Department
of Biochemistry, University of Puerto Rico, Medical Science Campus, San Juan 00921, Puerto Rico
| | - Carmen Acosta
- Department
of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan 00931, Puerto Rico
| | - Emily M. Ormé
- Department
of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan 00931, Puerto Rico
| | - Israel Rodríguez
- Department
of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan 00931, Puerto Rico
| | - Sara M. Delgado-Rivera
- Department
of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan 00931, Puerto Rico
| | - Arthur D. Tinoco
- Department
of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan 00931, Puerto Rico
| | - Suranganie Dharmawardhane
- Department
of Biochemistry, University of Puerto Rico, Medical Science Campus, San Juan 00921, Puerto Rico
| | | |
Collapse
|
3
|
Rampogu S, Balasubramaniyam T, Lee JH. Curcumin Chalcone Derivatives Database (CCDD): a Python framework for natural compound derivatives database. PeerJ 2023; 11:e15885. [PMID: 37605747 PMCID: PMC10440061 DOI: 10.7717/peerj.15885] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/20/2023] [Indexed: 08/23/2023] Open
Abstract
We built the Curcumin Chalcone Derivatives Database (CCDD) to enable the effective virtual screening of highly potent curcumin and its analogs. The two-dimensional (2D) structures were drawn using the ChemBioOffice package and converted to 3D structures using Discovery Studio Visualizer V 2021 (DS). The database was built using different Python modules. For the 3D structures, different Python packages were used to obtain the data frame of compounds. This framework is also used to visualize the compounds. The webserver enables the users to screen the compounds according to Lipinski's rule of five. The structures can be downloaded in .sdf and .mol format. The data frame (df) can be downloaded in .csv format. Our webserver can help computational drug discovery researchers find new therapeutics and build new webservers. The CCDD is freely available at: https://srampogu-ccdd-ccdd-8uldk8.streamlit.app/.
Collapse
Affiliation(s)
| | | | - Joon-Hwa Lee
- Department of Chemistry, Gyeongsang National University, Jinju, Gyeongnam, South Korea
| |
Collapse
|
4
|
Targeting TRAIL Death Receptors in Triple-Negative Breast Cancers: Challenges and Strategies for Cancer Therapy. Cells 2022; 11:cells11233717. [PMID: 36496977 PMCID: PMC9739296 DOI: 10.3390/cells11233717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The tumor necrosis factor (TNF) superfamily member TNF-related apoptosis-inducing ligand (TRAIL) induces apoptosis in cancer cells via death receptor (DR) activation with little toxicity to normal cells or tissues. The selectivity for activating apoptosis in cancer cells confers an ideal therapeutic characteristic to TRAIL, which has led to the development and clinical testing of many DR agonists. However, TRAIL/DR targeting therapies have been widely ineffective in clinical trials of various malignancies for reasons that remain poorly understood. Triple negative breast cancer (TNBC) has the worst prognosis among breast cancers. Targeting the TRAIL DR pathway has shown notable efficacy in a subset of TNBC in preclinical models but again has not shown appreciable activity in clinical trials. In this review, we will discuss the signaling components and mechanisms governing TRAIL pathway activation and clinical trial findings discussed with a focus on TNBC. Challenges and potential solutions for using DR agonists in the clinic are also discussed, including consideration of the pharmacokinetic and pharmacodynamic properties of DR agonists, patient selection by predictive biomarkers, and potential combination therapies. Moreover, recent findings on the impact of TRAIL treatment on the immune response, as well as novel strategies to address those challenges, are discussed.
Collapse
|
5
|
Sireesha R, Tej MB, Poojith N, Sreenivasulu R, Musuluri M, Subbarao M. Synthesis of Substituted Aryl Incorporated Oxazolo[4,5-b]Pyridine-Triazole Derivatives: Anticancer Evaluation and Molecular Docking Studies. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.2021256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Reddymasu Sireesha
- Department of Chemistry, Acharya Nagarjuna University, Guntur, Andhra Pradesh, India
| | - Mandava Bhuvan Tej
- Department of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamilnadu, India
| | | | - Reddymasu Sreenivasulu
- Department of Chemistry, University College of Engineering (Autonomous), Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh, India
| | - Murali Musuluri
- Department of Chemistry, RVR & JC College of Engineering, Guntur, Andhra Pradesh, India
| | - Mannam Subbarao
- Department of Chemistry, Acharya Nagarjuna University, Guntur, Andhra Pradesh, India
| |
Collapse
|
6
|
Gao F, Huang G, Xiao J. Chalcone hybrids as potential anticancer agents: Current development, mechanism of action, and structure-activity relationship. Med Res Rev 2020; 40:2049-2084. [PMID: 32525247 DOI: 10.1002/med.21698] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022]
Abstract
The continuous emergency of drug-resistant cancers and the low specificity of anticancer agents have been the major challenges in the control and treatment of cancer, making an urgent need to develop novel anticancer agents with high efficacy. Chalcones, precursors of flavonoids and isoflavonoids, exhibit structural heterogeneity and can act on various drug targets. Chalcones which demonstrated potential in vitro and in vivo activity against both drug-susceptible and drug-resistant cancers, are useful templates for the development of novel anticancer agents. Hybridization of chalcone moiety with other anticancer pharmacophores could provide the hybrids which have the potential to overcome drug resistance and improve the specificity, so it represents a promising strategy to develop novel anticancer agents. This review emphasizes the development, the mechanisms of action as well as structure-activity relationships of chalcone hybrids with potential therapeutic application for many cancers in recent 10 years.
Collapse
Affiliation(s)
- Feng Gao
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jiaqi Xiao
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
7
|
Eghbaliferiz S, Farhadi F, Barreto GE, Majeed M, Sahebkar A. Effects of curcumin on neurological diseases: focus on astrocytes. Pharmacol Rep 2020; 72:769-782. [PMID: 32458309 DOI: 10.1007/s43440-020-00112-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Astrocytes are the most abundant glial cells in the central nervous system, and are important players in both brain injury and neurodegenerative disease. Curcumin (1,7-bis[4-hydroxy-3-methoxyphenyl]-1,6-heptadiene-3,5-dione), the major active component of turmeric, belongs to the curcuminoid family that was originally isolated from the plant Curcuma longa. Several studies suggest that curcumin may have a beneficial impact on the brain pathology and aging. These effects are due to curcumin's antioxidant, free-radical scavenging, and anti-inflammatory activity. In light of this, our current review aims to discuss the role of astrocytes as essential players in neurodegenerative diseases and suggest that curcumin is capable of direct inhibition of astrocyte activity with a particular focus on its effects in Alexander disease, Alzheimer's disease, ischemia stroke, spinal cord injury, Multiple sclerosis, and Parkinson's disease.
Collapse
Affiliation(s)
- Samira Eghbaliferiz
- Department of Pharmacognosy, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Faegheh Farhadi
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | | | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Rodrigues FC, Anil Kumar NV, Thakur G. Developments in the anticancer activity of structurally modified curcumin: An up-to-date review. Eur J Med Chem 2019; 177:76-104. [PMID: 31129455 DOI: 10.1016/j.ejmech.2019.04.058] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/14/2019] [Accepted: 04/20/2019] [Indexed: 01/10/2023]
Abstract
Curcumin is a pharmacologically active polyphenol derived from the popular spice element-Turmeric. The therapeutic activity of curcumin has been extensively investigated over the last few decades and reports suggest the role of curcumin in a large number of biological activities, particularly its prominent anticancer activity. Curcumin, being a pleiotropic molecule, is a regulator of multiple molecular targets which play crucial roles in various cell signaling pathways. It is known to suppress transformation, inhibit proliferation as well as induce apoptosis. However, despite all these benefits, the efficacy of curcumin remains limited due to its poor bioavailability, poor absorption within the systemic circulation and rapid elimination from the body. To overcome these limiting factors, researchers all around the world are working towards designing a synthetic and superior curcuminoid by making suitable structural modifications to the parent skeleton. These curcuminoids, mainly analogues and derivatives, will not only improve the physicochemical properties but also enhance the efficacy simultaneously. The present review will provide a comprehensive account of the analogues and derivatives of curcumin that have been reported since 2014 which have indicated a better anticancer activity than curcumin.
Collapse
Affiliation(s)
- Fiona C Rodrigues
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, India
| | - N V Anil Kumar
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, India
| | - Goutam Thakur
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, India.
| |
Collapse
|
9
|
Diethylamino-curcumin mimic with trizolyl benzene enhances TRAIL-mediated cell death on human glioblastoma cells. Mol Cell Toxicol 2018. [DOI: 10.1007/s13273-018-0026-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
10
|
Celik H, Aydin T, Solak K, Khalid S, Farooqi AA. Curcumin on the "flying carpets" to modulate different signal transduction cascades in cancers: Next-generation approach to bridge translational gaps. J Cell Biochem 2018; 119:4293-4303. [PMID: 29384224 DOI: 10.1002/jcb.26749] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/25/2018] [Indexed: 12/25/2022]
Abstract
Curcumin, a bioactive and pharmacologically efficient component isolated from Curcuma longa has attracted considerable attention because of its ability to modulate diverse cellular and physiological pathways. WNT, TGF/SMAD, NOTCH, and SHH are fundamentally different signaling cascades, but their choreographed activation is strongly associated with cancer development and progression. In this review we have attempted to set spotlight on regulation of different cell signaling pathways by curcumin in different cancers. We partition this multi-component review into in-depth biological understanding of various signal transduction cascades and how curcumin targets intracellular signal transducers of deregulated pathways to inhibit cancer development and progression. Rapidly broadening landscape of both established and candidate oncogenic driver mutations identified in different cancers is a major stumbling block in the standardization of drugs having significant clinical outcome. Intra and inter-tumor heterogeneity had leveraged the complexity of therapeutic challenges to another level. Multi-pronged approach and molecularly guided treatments will be helpful in improving the clinical outcome.
Collapse
Affiliation(s)
- Hulya Celik
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Agri Ibrahim Cecen University, Agri, Turkey
| | - Tuba Aydin
- Department of Pharmacognosy, Faculty of Pharmacy, Agri Ibrahim Cecen University, Agri, Turkey
| | - Kubra Solak
- Institute of Science, Ataturk University, Erzurum, Turkey
| | - Sumbul Khalid
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Ammad A Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| |
Collapse
|
11
|
Yang X, Li Z, Wu Q, Chen S, Yi C, Gong C. TRAIL and curcumin codelivery nanoparticles enhance TRAIL-induced apoptosis through upregulation of death receptors. Drug Deliv 2017; 24:1526-1536. [PMID: 28994313 PMCID: PMC8241104 DOI: 10.1080/10717544.2017.1384863] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/22/2017] [Accepted: 09/22/2017] [Indexed: 02/05/2023] Open
Abstract
Active targeting nanoparticles were developed to simultaneously codeliver tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and Curcumin (Cur). In the nanoparticles (TRAIL-Cur-NPs), TRAIL was used as both active targeting ligand and therapeutic agent, and Cur could upregulate death receptors (DR4 and DR5) to increase the apoptosis-inducing effects of TRAIL. Compared with corresponding free drugs, TRAIL-Cur-NPs group showed enhanced cellular uptake, cytotoxicity and apoptosis induction effect on HCT116 colon cancer cells. In addition, in vivo anticancer studies suggested that TRAIL-Cur-NPs had superior therapeutic effect on tumors without obvious toxicity, which was mainly due to the high tumor targeting and synergistic effect of TRAIL and Cur. The synergistic mechanism of improved antitumor efficacy was proved to be upregulation of DR4 and DR5 in tumor cells induced by Cur. Thus, the prepared codelivery nanoparticles may have potential applications in colorectal cancer therapy.
Collapse
Affiliation(s)
- Xi Yang
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhaojun Li
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiotherapy, Hainan General Hospital, Haikou, China
| | - Qinjie Wu
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shouchun Chen
- Chengdu Huachuang Biotechnology Co. Ltd, Chengdu, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Changyang Gong
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Kang X, Zhao C, Yan L, Qi R, Jing X, Wang Z. Sensitizing nanoparticle based platinum(IV) drugs by curcumin for better chemotherapy. Colloids Surf B Biointerfaces 2016; 145:812-819. [DOI: 10.1016/j.colsurfb.2016.05.084] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 05/11/2016] [Accepted: 05/28/2016] [Indexed: 11/30/2022]
|
13
|
Chalcone Scaffold in Anticancer Armamentarium: A Molecular Insight. J Toxicol 2016; 2016:7651047. [PMID: 26880913 PMCID: PMC4735904 DOI: 10.1155/2016/7651047] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/07/2015] [Accepted: 12/15/2015] [Indexed: 12/26/2022] Open
Abstract
Cancer is an inevitable matter of concern in the medicinal chemistry era. Chalcone is the well exploited scaffold in the anticancer domain. The molecular mechanism of chalcone at cellular level was explored in past decades. This mini review provides the most recent updates on anticancer potential of chalcones.
Collapse
|
14
|
Mahmood K, Zia KM, Zuber M, Salman M, Anjum MN. Recent developments in curcumin and curcumin based polymeric materials for biomedical applications: A review. Int J Biol Macromol 2015; 81:877-90. [PMID: 26391597 DOI: 10.1016/j.ijbiomac.2015.09.026] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 09/05/2015] [Accepted: 09/16/2015] [Indexed: 01/15/2023]
Abstract
Turmeric (Curcuma longa) is a popular Indian spice that has been used for centuries in herbal medicines for the treatment of a variety of ailments such as rheumatism, diabetic ulcers, anorexia, cough and sinusitis. Curcumin (diferuloylmethane) is the main curcuminoid present in turmeric and responsible for its yellow color. Curcumin has been shown to possess significant anti-inflammatory, anti-oxidant, anti-carcinogenic, anti-mutagenic, anticoagulant and anti-infective effects. This review summarizes and discusses recently published papers on the key biomedical applications of curcumin based materials. The highlighted studies in the review provide evidence of the ability of curcumin to show the significant vitro antioxidant, diabetic complication, antimicrobial, neuroprotective, anti-cancer activities and detection of hypochlorous acid, wound healing, treatment of major depression, healing of paracentesis, and treatment of carcinoma and optical detection of pyrrole properties. Hydrophobic nature of this polyphenolic compound along with its rapid metabolism, physicochemical and biological instability contribute to its poor bioavailability. To redress these problems several approaches have been proposed like encapsulation of curcumin in liposomes and polymeric micelles, inclusion complex formation with cyclodextrin, formation of polymer-curcumin conjugates, etc.
Collapse
Affiliation(s)
- Kashif Mahmood
- Institute of Chemistry, Government College University, Faisalabad, Pakistan
| | - Khalid Mahmood Zia
- Institute of Chemistry, Government College University, Faisalabad, Pakistan.
| | - Mohammad Zuber
- Institute of Chemistry, Government College University, Faisalabad, Pakistan
| | - Mahwish Salman
- Institute of Chemistry, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
15
|
Park CB, Ahn CM, Oh S, Kwon D, Cho WC, Shin WS, Cui Y, Um YS, Park BG, Lee S. Synthesis of alkylsulfonyl and substituted benzenesulfonyl curcumin mimics as dual antagonist of L-type Ca(2+) channel and endothelin A/B2 receptor. Bioorg Med Chem 2015; 23:6673-82. [PMID: 26386817 DOI: 10.1016/j.bmc.2015.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/03/2015] [Accepted: 09/04/2015] [Indexed: 12/01/2022]
Abstract
We synthesized a library of curcumin mimics with diverse alkylsulfonyl and substituted benzenesulfonyl modifications through a simple addition reaction of important intermediate, 1-(3-Amino-phenyl)-3-(4-hydroxy-3-methoxy-phenyl)-propenone (10), with various sulfonyl chloride reactants and then tested their vasodilatation effect on depolarization (50 mM K(+))- and endothelin-1 (ET-1)-induced basilar artery contraction. Generally, curcumin mimics with aromatic sulfonyl groups showed stronger vasodilation effect than alkyl sulfonylated curcumin mimics. Among the tested compounds, six curcumin mimics (11g, 11h, 11i, 11j, 11l, and 11s) in a depolarization-induced vasoconstriction and seven compounds (11g, 11h, 11i, 11j, 11l, 11p, and 11s) in an ET-1-induced vasoconstriction showed strong vasodilation effect. Based on their biological properties, synthetic curcumin mimics can act as dual antagonist scaffold of L-type Ca(2+) channel and endothelin A/B2 receptor in vascular smooth muscle cells. In particular, compounds 11g and 11s are promising novel drug candidates to treat hypertension related to the overexpression of L-type Ca(2+) channels and ET peptides/receptors-mediated cardiovascular diseases.
Collapse
Affiliation(s)
- Chong-Bin Park
- Department of Thoracic and Cardiovascular Surgery, Gangneung Asan Hospital, Ulsan University College of Medicine, Gangneung 210-711, Republic of Korea
| | - Chan Mug Ahn
- Department of Basic Science, Yonsei University Wonju College of Medicine, Wonju 220-701, Republic of Korea
| | - Sangtae Oh
- Department of Basic Science, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea
| | - Daeho Kwon
- Department of Microbiology, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea; Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea
| | - Won-Chul Cho
- Department of Thoracic and Cardiovascular Surgery, Gangneung Asan Hospital, Ulsan University College of Medicine, Gangneung 210-711, Republic of Korea
| | - Woon-Seob Shin
- Department of Microbiology, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea; Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea
| | - Yuan Cui
- Department of Physiology, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea
| | - Ye Sol Um
- Department of Physiology, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea
| | - Byong-Gon Park
- Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea; Department of Physiology, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea.
| | - Seokjoon Lee
- Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea; Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 210-701, Republic of Korea.
| |
Collapse
|