1
|
Wang S, Wang J, Lu X, Liu M, Liu Y, Li M, Kong X, Wu L, Guan Q, Zhang W. Design, synthesis and biological evaluation of novel tubulin-targeting agents with a dual-mechanism for polymerization inhibition and protein degradation. Eur J Med Chem 2024; 272:116458. [PMID: 38703557 DOI: 10.1016/j.ejmech.2024.116458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/12/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024]
Abstract
Microtubules are recognized as one of the most vital and attractive targets in anticancer therapy. The development of novel tubulin-targeting agents with a new action mechanism is imperative. Based on the hydrophobic tagging strategy, the molecular scaffold of tirbanibulin was selected as tubulin target-binding moiety, subsequent to which a series of target compounds were rationally designed by selecting various combinations of linkers and hydrophobic tags. A set of novel molecules were synthesized and most of them exhibited potent antiproliferative activity against tumor cells in vitro. The most active compound 14b inhibited polymerization of purified recombinant tubulin and induced degradation of α- and β-tubulin in MCF-7 cells. Notably, following treatment with compound 14b, an unexpected phenomenon of "microtubules fragmentation" was observed via immunofluorescence staining. Furthermore, compound 14b possessed antitumor activity in the 4T1 allograft models with TGI of 74.27 % without significant toxicity. In this work, we report the discovery of novel dual-mechanism tubulin-targeting agents.
Collapse
Affiliation(s)
- Sibo Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Jiahao Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xiankun Lu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Meitong Liu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Yue Liu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Mi Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xuejie Kong
- Department of Geratology, The First Affiliated Hospital, Chinese Medical University, Shenyang, 110001, China
| | - Lan Wu
- Department of Geratology, The First Affiliated Hospital, Chinese Medical University, Shenyang, 110001, China.
| | - Qi Guan
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
2
|
Yang H, Qin J, Pei Y, Guan S, Zhao M, Wang Y, Yao Y, Duan Y, Sun M. Discovery of the cereblon-recruiting tubulin PROTACs effective in overcoming Taxol resistance in vitro and in vivo. Eur J Med Chem 2024; 265:116067. [PMID: 38171146 DOI: 10.1016/j.ejmech.2023.116067] [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: 10/26/2023] [Revised: 11/29/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
Overexpression of β3-tubulin is a common occurrence in human tumors and is associated with resistance to microtubule-targeting agents. PROTAC strategy has demonstrated significant potential in overcoming drug resistance. Herein, we report the discovery of W13 as the first PROTAC against tubulin, which was created by connecting a CRBN ligand to the widely recognized microtubule-destabilizing agent CA-4. Notably, it retains the inhibitory activity of the parental CA-4 and further exhibits substantial degradation of α/β/β3-tubulin in both A549 and A549/Taxol cell lines. The degradation of tubulin was subsequently verified to be mediated by the ubiquitin-proteasome system. Importantly, tumor xenograft research clearly showed W13's promising antitumor activity against human lung cancer. Taken together, the discovery of W13 demonstrated the practicality and feasibility of PROTAC targeting tubulin, hence establishing a potential therapeutic approach for treating NSCLC caused by the overexpression of β3-tubulin.
Collapse
Affiliation(s)
- Hua Yang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Jinling Qin
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yuanyuan Pei
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Sumeng Guan
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Mei Zhao
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yingge Wang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yongfang Yao
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China.
| | - Moran Sun
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
3
|
Padathpeedika Khalid J, Mary Martin T, Prathap L, Abhimanyu Nisargandha M, Boopathy N, Kishore Kumar MS. Exploring Tumor-Promoting Qualities of Cancer-Associated Fibroblasts and Innovative Drug Discovery Strategies With Emphasis on Thymoquinone. Cureus 2024; 16:e53949. [PMID: 38468988 PMCID: PMC10925941 DOI: 10.7759/cureus.53949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/09/2024] [Indexed: 03/13/2024] Open
Abstract
Tumor epithelial development and chemoresistance are highly promoted by the tumor microenvironment (TME), which is mostly made up of the cancer stroma. This is due to several causes. Cancer-associated fibroblasts (CAFs) stand out among them as being essential for the promotion of tumors. Understanding the fibroblastic population within a single tumor is made more challenging by the undeniable heterogeneity within it, even though particular stromal alterations are still up for debate. Numerous chemical signals released by tumors improve the connections between heterotypic fibroblasts and CAFs, promoting the spread of cancer. It becomes essential to have a thorough understanding of this complex microenvironment to effectively prevent solid tumor growth. Important new insights into the role of CAFs in the TME have been revealed by recent studies. The objective of this review is to carefully investigate the relationship between CAFs in tumors and plant secondary metabolites, with a focus on thymoquinone (TQ). The literature published between 2010 and 2023 was searched in PubMed and Google Scholar with keywords such as TQ, TME, cancer-associated fibroblasts, mechanism of action, and flavonoids. The results showed a wealth of data substantiating the activity of plant secondary metabolites, particularly TQ's involvement in blocking CAF operations. Scrutinized research also clarified the wider effect of flavonoids on pathways related to cancer. The present study highlights the complex dynamics of the TME and emphasizes the critical role of CAFs. It also examines the possible interventions provided by secondary metabolites found in plants, with TQ playing a vital role in regulating CAF function based on recent literature.
Collapse
Affiliation(s)
- Jabir Padathpeedika Khalid
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Taniya Mary Martin
- Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Lavanya Prathap
- Department of Anatomy, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Milind Abhimanyu Nisargandha
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Nisha Boopathy
- Department of Community Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Meenakshi Sundaram Kishore Kumar
- Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
4
|
Cui Y, Zhang J, Zhang G. The Potential Strategies for Overcoming Multidrug Resistance and Reducing Side Effects of Monomer Tubulin Inhibitors for Cancer Therapy. Curr Med Chem 2024; 31:1874-1895. [PMID: 37349994 DOI: 10.2174/0929867330666230622142505] [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: 01/29/2023] [Revised: 04/26/2023] [Accepted: 05/12/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Tubulin is an essential target in tumor therapy, and this is attributed to its ability to target MT dynamics and interfere with critical cellular functions, including mitosis, cell signaling, and intracellular trafficking. Several tubulin inhibitors have been approved for clinical application. However, the shortcomings, such as drug resistance and toxic side effects, limit its clinical application. Compared with single-target drugs, multi-target drugs can effectively improve efficacy to reduce side effects and overcome the development of drug resistance. Tubulin protein degraders do not require high concentrations and can be recycled. After degradation, the protein needs to be resynthesized to regain function, which significantly delays the development of drug resistance. METHODS Using SciFinder® as a tool, the publications about tubulin-based dual-target inhibitors and tubulin degraders were surveyed with an exclusion of those published as patents. RESULTS This study presents the research progress of tubulin-based dual-target inhibitors and tubulin degraders as antitumor agents to provide a reference for developing and applying more efficient drugs for cancer therapy. CONCLUSION The multi-target inhibitors and protein degraders have shown a development prospect to overcome multidrug resistance and reduce side effects in the treatment of tumors. Currently, the design of dual-target inhibitors for tubulin needs to be further optimized, and it is worth further clarifying the detailed mechanism of protein degradation.
Collapse
Affiliation(s)
- Yingjie Cui
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, P.R. China
| | - Jing Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, P.R. China
| | - Guifang Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, P.R. China
| |
Collapse
|
5
|
Sheikhnia F, Rashidi V, Maghsoudi H, Majidinia M. Potential anticancer properties and mechanisms of thymoquinone in colorectal cancer. Cancer Cell Int 2023; 23:320. [PMID: 38087345 PMCID: PMC10717210 DOI: 10.1186/s12935-023-03174-4] [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: 05/25/2023] [Accepted: 12/04/2023] [Indexed: 10/14/2024] Open
Abstract
Colorectal neoplasms are one of the deadliest diseases among all cancers worldwide. Thymoquinone (TQ) is a natural compound of Nigella sativa that has been used in traditional medicine against a variety of acute/chronic diseases such as asthma, bronchitis, rheumatism, headache, back pain, anorexia, amenorrhea, paralysis, inflammation, mental disability, eczema, obesity, infections, depression, dysentery, hypertension, gastrointestinal, cardiovascular, hepatic, and renal disorders. This review aims to present a detailed report on the studies conducted on the anti-cancer properties of TQ against colorectal cancer, both in vitro and in vivo. TQ stands as a promising natural therapeutic agent that can enhance the efficacy of existing cancer treatments while minimizing the associated adverse effects. The combination of TQ with other anti-neoplastic agents promoted the efficacy of existing cancer treatments. Further research is needed to acquire a more comprehensive understanding of its exact molecular targets and pathways and maximize its clinical usefulness. These investigations may potentially aid in the development of novel techniques to combat drug resistance and surmount the obstacles presented by chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Farhad Sheikhnia
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Vahid Rashidi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hossein Maghsoudi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
6
|
Song Y, Liu H, Xian Q, Gui C, Xu M, Zhou Y. Mechanistic insights into UHRF1‑mediated DNA methylation by structure‑based functional clarification of UHRF1 domains (Review). Oncol Lett 2023; 26:542. [PMID: 38020304 PMCID: PMC10660443 DOI: 10.3892/ol.2023.14129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Epigenetic modification is crucial for transmitting genetic information, while abnormalities in DNA methylation modification are primarily associated with cancer and neurological diseases. As a multifunctional epigenetic modifier, ubiquitin like with PHD and ring finger domains 1 (UHRF1) mainly affects cell energy metabolism and cell cycle control. It also inhibits the transcription of tumor suppressor genes through DNA and/or histone methylation modifications, promoting the occurrence and development of cancer. Therefore, comprehensively understanding the molecular mechanism of the epigenetic modification of UHRF1 in tumors will help identify targets for inhibiting the expression and function of UHRF1. Notably, each domain of UHRF1 functions as a whole and differently. Thus, the abnormality of any domain can lead to a change in phenotype or disease. However, the specific regulatory mechanism and proteins of each domain have not been fully elucidated. The present review aimed to contribute to the study of the regulatory mechanism of UHRF1 to a greater extent in different cancers and provide ideas for drug research by clarifying the function of UHRF1 domains.
Collapse
Affiliation(s)
- Yiying Song
- Department of Clinical Laboratory Diagnosis, Jinan Central Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Haiting Liu
- Department of Critical Care Medicine, Jinan Zhangqiu Hospital of Traditional Chinese Medicine, Jinan, Shandong 250200, P.R. China
| | - Qingqing Xian
- Department of Clinical Laboratory Diagnosis, Jinan Central Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Chengzhi Gui
- Department of Clinical Laboratory Diagnosis, Shandong First Medical University, Jinan, Shandong 250012, P.R. China
| | - Mingjie Xu
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| | - Yunying Zhou
- Department of Clinical Laboratory Diagnosis, Jinan Central Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
- Department of Clinical Laboratory Diagnosis, Shandong First Medical University, Jinan, Shandong 250012, P.R. China
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
7
|
Abstract
Targeted protein degradation (TPD) has emerged as the most promising approach for the specific knockdown of disease-associated proteins and is achieved by exploiting the cellular quality control machinery. TPD technologies are highly advantageous in overcoming drug resistance as they degrade the whole target protein. Microtubules play important roles in many cellular processes and are among the oldest and most well-established targets for tumor chemotherapy. However, the development of drug resistance, risk of hypersensitivity reactions, and intolerable toxicities severely restrict the clinical applications of microtubule-targeting agents (MTAs). Microtubule degradation agents (MDgAs) operate via completely different mechanisms compared with traditional MTAs and are capable of overcoming drug resistance. The emergence of MDgAs has expanded the scope of TPD and provided new avenues for the discovery of tubulin-targeted drugs. Herein, we summarized the development of MDgAs, and discussed their degradation mechanisms, mechanisms of action on the binding sites, potential opportunities, and challenges.
Collapse
Affiliation(s)
- Chufeng Zhang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Min Zhao
- Laboratory of Metabolomics and Drug-Induced Liver Injury, Department of Gastroenterology & Hepatology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yong Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
8
|
Sen S, Kasikci M. Low-dose rosmarinic acid and thymoquinone accelerate wound healing in retinal pigment epithelial cells. Int Ophthalmol 2023; 43:3811-3821. [PMID: 37407754 DOI: 10.1007/s10792-023-02799-8] [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: 05/03/2023] [Accepted: 06/22/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE Thymoquinone (TQ) and rosmarinic acid (RA) are two biologically active compounds found in plants and that possess remarkable anti-oxidant and anti-inflammatory properties. The present study aimed to investigate the potential protective effects of RA and TQ, which have known anti-inflammatory and anti-oxidant effects, on retinal damage by establishing a wound healing model for retinal pigment epithelial cells (ARPE-19). METHOD To this end, IC50 doses of RA and TQ in ARPE-19 cells were calculated by MTT assay. Both agents were administered at IC50, IC50/2 and IC50/4 doses for wound healing assay, and wound closure percentages were analyzed. Since the best wound healing was found at IC50/4 dose (low dose) for both agents, other biochemical and molecular analyses were planned to be performed using these doses. Following low dose RA and TQ treatments, the cells were lysed and TGF-β1 and MMP-9 levels were analyzed by ELISA technique from the cell lysates obtained. In addition, the mRNA expression levels of TLR3, IFN-γ and VEGF were calculated by RT-PCR technique. RESULTS Low dose of RA and TQ dramatically increased wound healing. RA may have achieved this by increasing levels of MMP-9 and TLR-3. In contrast, the mRNA expression level of VEGF remained unchanged. TQ accelerated wound healing by increasing both the protein levels of TGF-β1 and MMP-9. Furthermore, low dose of TQ decreased both TLR3 and IFN-γ mRNA expression levels. CONCLUSION Low doses of RA and TQ were clearly demonstrated to have protective properties against possible damage to retinal pigment epithelial cells.
Collapse
Affiliation(s)
- Serkan Sen
- Department of Medical Laboratory Techniques, Ataturk Vocational School of Health Services, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
- Department of Ophthalmology, Mugla Education and Research Hospital, Mugla, Turkey
| | - Murat Kasikci
- Department of Medical Laboratory Techniques, Ataturk Vocational School of Health Services, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey.
- Department of Ophthalmology, Mugla Education and Research Hospital, Mugla, Turkey.
| |
Collapse
|
9
|
Zhang YF, Huang J, Zhang WX, Liu YH, Wang X, Song J, Jin CY, Zhang SY. Tubulin degradation: Principles, agents, and applications. Bioorg Chem 2023; 139:106684. [PMID: 37356337 DOI: 10.1016/j.bioorg.2023.106684] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023]
Abstract
The microtubule system plays an important role in the mitosis and growth of eukaryotic cells, and it is considered as an appealing and highly successful molecular target for cancer treatment. In fact, microtubule targeting agents, such as paclitaxel and vinblastine, have been approved by FDA for tumor therapy, which have achieved significant therapeutic effects and sales performance. At present, microtubule targeting agents mainly include microtubule-destabilizing agents, microtubule-stabilizing agents, and a few tubulin degradation agents. Although there are few reports about tubulin degradation agents at present, tubulin degradation agents show great potential in overcoming multidrug resistance and reducing neurotoxicity. In addition, some natural drugs could specifically degrade tubulin in tumor cells, but have no effect in normal cells, thus showing a good biosafety profile. Therefore, tubulin degradation agents might exhibit a better application. Currently, some small molecules have been designed to promote tubulin degradation with potent antiproliferative activities, showing the potential for cancer treatment. In this work, we reviewed the reports on tubulin degradation, and focused on the degradation mechanism and important functional groups of chemically synthesized compounds, hoping to provide help for the degradation design of tubulin.
Collapse
Affiliation(s)
- Yi-Fan Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China
| | - Jiao Huang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China
| | - Wei-Xin Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China
| | - Yun-He Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China
| | - Xiao Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jian Song
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Cheng-Yun Jin
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China.
| | - Sai-Yang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
10
|
Tabassum S, Thakur V, Rosli N, Ichwan SJA, Mishra P, Suriyah WH. Therapeutic implications of thymoquinone and its molecular and functional mechanisms against oral and lung cancer. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
11
|
Therapeutic Potential of Certain Terpenoids as Anticancer Agents: A Scoping Review. Cancers (Basel) 2022; 14:cancers14051100. [PMID: 35267408 PMCID: PMC8909202 DOI: 10.3390/cancers14051100] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is a life-threatening disease and is considered to be among the leading causes of death worldwide. Chemoresistance, severe toxicity, relapse and metastasis are the major obstacles in cancer therapy. Therefore, introducing new therapeutic agents for cancer remains a priority to increase the range of effective treatments. Terpenoids, a large group of secondary metabolites, are derived from plant sources and are composed of several isoprene units. The high diversity of terpenoids has drawn attention to their potential anticancer and pharmacological activities. Some terpenoids exhibit an anticancer effect by triggering various stages of cancer progression, for example, suppressing the early stage of tumorigenesis via induction of cell cycle arrest, inhibiting cancer cell differentiation and activating apoptosis. At the late stage of cancer development, certain terpenoids are able to inhibit angiogenesis and metastasis via modulation of different intracellular signaling pathways. Significant progress in the identification of the mechanism of action and signaling pathways through which terpenoids exert their anticancer effects has been highlighted. Hence, in this review, the anticancer activities of twenty-five terpenoids are discussed in detail. In addition, this review provides insights on the current clinical trials and future directions towards the development of certain terpenoids as potential anticancer agents.
Collapse
|
12
|
Li Y, Liu Y, Zhu Z, Yan W, Zhang C, Yang Z, Bai P, Tang M, Shi M, He W, Fu S, Liu J, Han K, Li J, Xie L, Ye H, Yang J, Chen L. Structure-Based Design and Synthesis of N-Substituted 3-Amino-β-Carboline Derivatives as Potent αβ-Tubulin Degradation Agents. J Med Chem 2022; 65:2675-2693. [PMID: 35084853 DOI: 10.1021/acs.jmedchem.1c02159] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
So far, relatively few small molecules have been reported to promote tubulin degradation. Our previous studies have found that compound 2, a noncovalent colchicine-site ligand, was capable of promoting αβ-tubulin degradation. To further improve its antiproliferative activity, 66 derivatives or analogues of 2 were designed and synthesized based on 2-tubulin cocrystal structure. Among them, 12b displayed nanomolar potency against a variety of tumor cells, including paclitaxel- and adriamycin-resistant cell lines. 12b binds to the colchicine site and promotes αβ-tubulin degradation in a concentration-dependent manner via the ubiquitin-proteasome pathway. The X-ray crystal structure revealed that 12b binds in a similar manner as 2, but there is a slight conformation change of the B ring, which resulted in better interaction of 12b with surrounding residues. 12b effectively suppressed tumor growth at an i.v. dose of 40 mg/kg (3 times a week) on both A2780S (paclitaxel-sensitive) and A2780T (paclitaxel-resistant) ovarian xenograft models, with respective TGIs of 92.42 and 79.75% without obvious side effects, supporting its potential utility as a tumor-therapeutic compound.
Collapse
Affiliation(s)
- Yong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Yan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zejiang Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Chufeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Peng Bai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Mingsong Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Wen He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Suhong Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Jiang Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Kai Han
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Jiewen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Lixin Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| |
Collapse
|
13
|
Medeiros M, Candido MF, Valera ET, Brassesco MS. The multifaceted NF-kB: are there still prospects of its inhibition for clinical intervention in pediatric central nervous system tumors? Cell Mol Life Sci 2021; 78:6161-6200. [PMID: 34333711 PMCID: PMC11072991 DOI: 10.1007/s00018-021-03906-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022]
Abstract
Despite advances in the understanding of the molecular mechanisms underlying the basic biology and pathogenesis of pediatric central nervous system (CNS) malignancies, patients still have an extremely unfavorable prognosis. Over the years, a plethora of natural and synthetic compounds has emerged for the pharmacologic intervention of the NF-kB pathway, one of the most frequently dysregulated signaling cascades in human cancer with key roles in cell growth, survival, and therapy resistance. Here, we provide a review about the state-of-the-art concerning the dysregulation of this hub transcription factor in the most prevalent pediatric CNS tumors: glioma, medulloblastoma, and ependymoma. Moreover, we compile the available literature on the anti-proliferative effects of varied NF-kB inhibitors acting alone or in combination with other therapies in vitro, in vivo, and clinical trials. As the wealth of basic research data continues to accumulate, recognizing NF-kB as a therapeutic target may provide important insights to treat these diseases, hopefully contributing to increase cure rates and lower side effects related to therapy.
Collapse
Affiliation(s)
- Mariana Medeiros
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - María Sol Brassesco
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, FFCLRP-USP, University of São Paulo, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, São Paulo, CEP 14040-901, Brazil.
| |
Collapse
|
14
|
A Fast Ubiquitination of UHRF1 Oncogene Is a Unique Feature and a Common Mechanism of Thymoquinone in Cancer Cells. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11167633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Downregulation of the ubiquitin-like containing PHD and ring finger 1 (UHRF1) oncogene in cancer cells in response to natural anticancer drugs, including thymoquinone (TQ), is a key event that induces apoptosis. TQ can induce UHRF1 autoubiquitination via the E3 ligase activity of its RING domain, most likely through the downregulation of herpes virus-associated ubiquitin-specific protease (HAUSP). In this study, we evaluated whether HAUSP downregulation and fast ubiquitination of UHRF1 are prerequisites for UHRF1 degradation in response to TQ in cancer cells and whether doxorubicin can mimic the effects of TQ on UHRF1 ubiquitination. RNA sequencing was performed to investigate differentially expressed genes in TQ-treated Jurkat cells. The protein expression of UHRF1, HAUSP and Bcl-2 was detected by means of Western blot analysis. The proliferation of human colon cancer (HCT-116) and Jurkat cells was analyzed via the WST-1 assay. RNA sequencing data revealed that TQ significantly decreased HAUSP expression. TQ triggered UHRF1 to undergo rapid ubiquitination as the first step in its degradation and the inhibition of its cell proliferation. TQ-induced UHRF1 ubiquitination is associated with HAUSP downregulation. Like TQ, doxorubicin induced a similar dose- and time-dependent downregulation of UHRF1 in cancer cells, but UHRF1 did not undergo ubiquitination as detected in response to TQ. Furthermore, TQ decreased Bcl-2 expression without triggering its ubiquitination. A fast UHRF1 ubiquitination is an indispensable event for its degradation in response to TQ but not for its responses to doxorubicin. TQ appears to trigger ubiquitination of UHRF1 but not of the Bcl-2 oncogene, thereby identifying UHRF1 as a specific target of TQ for cancer therapy.
Collapse
|
15
|
Abdullah O, Omran Z, Hosawi S, Hamiche A, Bronner C, Alhosin M. Thymoquinone Is a Multitarget Single Epidrug That Inhibits the UHRF1 Protein Complex. Genes (Basel) 2021; 12:genes12050622. [PMID: 33922029 PMCID: PMC8143546 DOI: 10.3390/genes12050622] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Silencing of tumor suppressor genes (TSGs) through epigenetic mechanisms, mainly via abnormal promoter DNA methylation, is considered a main mechanism of tumorigenesis. The abnormal DNA methylation profiles are transmitted from the cancer mother cell to the daughter cells through the involvement of a macromolecular complex in which the ubiquitin-like containing plant homeodomain (PHD), and an interesting new gene (RING) finger domains 1 (UHRF1), play the role of conductor. Indeed, UHRF1 interacts with epigenetic writers, such as DNA methyltransferase 1 (DNMT1), histone methyltransferase G9a, erasers like histone deacetylase 1 (HDAC1), and functions as a hub protein. Thus, targeting UHRF1 and/or its partners is a promising strategy for epigenetic cancer therapy. The natural compound thymoquinone (TQ) exhibits anticancer activities by targeting several cellular signaling pathways, including those involving UHRF1. In this review, we highlight TQ as a potential multitarget single epidrug that functions by targeting the UHRF1/DNMT1/HDAC1/G9a complex. We also speculate on the possibility that TQ might specifically target UHRF1, with subsequent regulatory effects on other partners.
Collapse
Affiliation(s)
- Omeima Abdullah
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (O.A.); (Z.O.)
| | - Ziad Omran
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (O.A.); (Z.O.)
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Ali Hamiche
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Université de Strasbourg, 67404 Illkirch, France; (A.H.); (C.B.)
| | - Christian Bronner
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Université de Strasbourg, 67404 Illkirch, France; (A.H.); (C.B.)
| | - Mahmoud Alhosin
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Correspondence: ; Tel.: +966-597-959-354
| |
Collapse
|
16
|
Omran Z, H. Dalhat M, Abdullah O, Kaleem M, Hosawi S, A Al-Abbasi F, Wu W, Choudhry H, Alhosin M. Targeting Post-Translational Modifications of the p73 Protein: A Promising Therapeutic Strategy for Tumors. Cancers (Basel) 2021; 13:cancers13081916. [PMID: 33921128 PMCID: PMC8071514 DOI: 10.3390/cancers13081916] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 01/11/2023] Open
Abstract
The tumor suppressor p73 is a member of the p53 family and is expressed as different isoforms with opposing properties. The TAp73 isoforms act as tumor suppressors and have pro-apoptotic effects, whereas the ΔNp73 isoforms lack the N-terminus transactivation domain and behave as oncogenes. The TAp73 protein has a high degree of similarity with both p53 function and structure, and it induces the regulation of various genes involved in the cell cycle and apoptosis. Unlike those of the p53 gene, the mutations in the p73 gene are very rare in tumors. Cancer cells have developed several mechanisms to inhibit the activity and/or expression of p73, from the hypermethylation of its promoter to the modulation of the ratio between its pro- and anti-apoptotic isoforms. The p73 protein is also decorated by a panel of post-translational modifications, including phosphorylation, acetylation, ubiquitin proteasomal pathway modifications, and small ubiquitin-related modifier (SUMO)ylation, that regulate its transcriptional activity, subcellular localization, and stability. These modifications orchestrate the multiple anti-proliferative and pro-apoptotic functions of TAp73, thereby offering multiple promising candidates for targeted anti-cancer therapies. In this review, we summarize the current knowledge of the different pathways implicated in the regulation of TAp73 at the post-translational level. This review also highlights the growing importance of targeting the post-translational modifications of TAp73 as a promising antitumor strategy, regardless of p53 status.
Collapse
Affiliation(s)
- Ziad Omran
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (Z.O.); (O.A.)
| | - Mahmood H. Dalhat
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Omeima Abdullah
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (Z.O.); (O.A.)
| | - Mohammed Kaleem
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Salman Hosawi
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Fahd A Al-Abbasi
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Wei Wu
- Department of Medicine, University of California, San Francisco, CA 94143, USA;
| | - Hani Choudhry
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
| | - Mahmoud Alhosin
- King Fahd Medical Research Center, Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.H.D.); (M.K.); (S.H.); (F.A.A.-A.); (H.C.)
- Correspondence: ; Tel.: +96-65-9795-9354
| |
Collapse
|
17
|
Ansary J, Giampieri F, Forbes-Hernandez TY, Regolo L, Quinzi D, Gracia Villar S, Garcia Villena E, Tutusaus Pifarre K, Alvarez-Suarez JM, Battino M, Cianciosi D. Nutritional Value and Preventive Role of Nigella sativa L. and Its Main Component Thymoquinone in Cancer: An Evidenced-Based Review of Preclinical and Clinical Studies. Molecules 2021; 26:molecules26082108. [PMID: 33916916 PMCID: PMC8067617 DOI: 10.3390/molecules26082108] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 11/16/2022] Open
Abstract
In recent times, scientific attention has been paid to different foods and their bioactive components for the ability to inhibit the onset and progress of different types of cancer. Nigella sativa extract, powder and seed oil and its main components, thymoquinone and α-hederin, have showed potent anticancer and chemosensitizing effects against various types of cancer, such as liver, colon, breast, renal, cervical, lung, ovarian, pancreatic, prostate and skin tumors, through the modulation of various molecular signaling pathways. Herein, the purpose of this review was to highlight the anticancer activity of Nigella sativa and it constitutes, focusing on different in vitro, in vivo and clinical studies and projects, in order to underline their antiproliferative, proapoptotic, cytotoxic and antimetastatic effects. Particular attention has been also given to the synergistic effect of Nigella sativa and it constitutes with chemotherapeutic drugs, and to the synthesized analogs of thymoquinone that seem to enhance the chemo-sensitizing potential. This review could be a useful step towards new research on N. sativa and cancer, to include this plant in the dietary treatments in support to conventional therapies, for the best achievement of therapeutic goals.
Collapse
Affiliation(s)
- Johura Ansary
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
| | - Francesca Giampieri
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Tamara Y. Forbes-Hernandez
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
| | - Lucia Regolo
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
| | - Denise Quinzi
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
| | - Santos Gracia Villar
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (S.G.V.); (E.G.V.); (K.T.P.)
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - Eduardo Garcia Villena
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (S.G.V.); (E.G.V.); (K.T.P.)
| | - Kilian Tutusaus Pifarre
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (S.G.V.); (E.G.V.); (K.T.P.)
- Research Center for Foods, Nutritional Biochemistry and Health, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - José M. Alvarez-Suarez
- Departamento de Ingeniería en Alimentos, Colegio de Ciencias e Ingenierías, Universidad San Francisco de Quito, Quito 170157, Ecuador
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (J.M.A.-S.); (M.B.); (D.C.); Tel.: +593-2-297-1700 (J.M.A.-S.); +339-071-220-4646 (M.B.); +339-071-220-4136 (D.C.)
| | - Maurizio Battino
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Correspondence: (J.M.A.-S.); (M.B.); (D.C.); Tel.: +593-2-297-1700 (J.M.A.-S.); +339-071-220-4646 (M.B.); +339-071-220-4136 (D.C.)
| | - Danila Cianciosi
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (J.A.); (F.G.); (L.R.); (D.Q.)
- Correspondence: (J.M.A.-S.); (M.B.); (D.C.); Tel.: +593-2-297-1700 (J.M.A.-S.); +339-071-220-4646 (M.B.); +339-071-220-4136 (D.C.)
| |
Collapse
|
18
|
Alhosin M, Razvi SSI, Sheikh RA, Khan JA, Zamzami MA, Choudhry H. Thymoquinone and Difluoromethylornithine (DFMO) Synergistically Induce Apoptosis of Human Acute T Lymphoblastic Leukemia Jurkat Cells Through the Modulation of Epigenetic Pathways. Technol Cancer Res Treat 2020; 19:1533033820947489. [PMID: 32912061 PMCID: PMC7488875 DOI: 10.1177/1533033820947489] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Thymoquinone (TQ), a natural anticancer agent exerts cytotoxic effects on several tumors by targeting multiple pathways, including apoptosis. Difluoromethylornithine (DFMO), an irreversible inhibitor of the ornithine decarboxylase (ODC) enzyme, has shown promising inhibitory activities in many cancers including leukemia by decreasing the biosynthesis of the intracellular polyamines. The present study aimed to investigate the combinatorial cytotoxic effects of TQ and DFMO on human acute T lymphoblastic leukemia Jurkat cells and to determine the underlying mechanisms. Here, we show that the combination of DFMO and TQ significantly reduced cell viability and resulted in significant synergistic effects on apoptosis when compared to either DFMO or TQ alone. RNA-sequencing showed that many key epigenetic players including Ubiquitin-like containing PHD and Ring finger 1 (UHRF1) and its 2 partners DNA methyltransferase 1 (DNMT1) and histone deacetylase 1 (HDAC1) were down-regulated in DFMO-treated Jurkat cells. The combination of DFMO and TQ dramatically decreased the expression of UHRF1, DNMT1 and HDAC1 genes compared to either DFMO or TQ alone. UHRF1 knockdown led to a decrease in Jurkat cell viability. In conclusion, these results suggest that the combination of DFMO and TQ could be a promising new strategy for the treatment of human acute T lymphoblastic leukemia by targeting the epigenetic code.
Collapse
Affiliation(s)
- Mahmoud Alhosin
- Department of Biochemistry, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer Metabolism and Epigenetic Unit, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| | - Syed Shoeb I Razvi
- Department of Biochemistry, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia.,Math and Science Department, 441417Community College of Qatar, Doha, Qatar
| | - Ryan A Sheikh
- Department of Biochemistry, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jalaluddin A Khan
- Department of Biochemistry, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mazin A Zamzami
- Department of Biochemistry, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer Metabolism and Epigenetic Unit, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer Metabolism and Epigenetic Unit, Faculty of Science, 37848King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
19
|
Duan Y, Liu W, Tian L, Mao Y, Song C. Targeting Tubulin-colchicine Site for Cancer Therapy: Inhibitors, Antibody- Drug Conjugates and Degradation Agents. Curr Top Med Chem 2019; 19:1289-1304. [PMID: 31210108 DOI: 10.2174/1568026619666190618130008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/22/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022]
Abstract
Microtubules are essential for the mitotic division of cells and have been an attractive target
for antitumour drugs due to the increased incidence of cancer and significant mitosis rate of tumour cells.
In the past few years, tubulin-colchicine binding site, as one of the three binding pockets including taxol-,
vinblastine- and colchicine-binding sites, has been focused on to design tubulin-destabilizing agents including
inhibitors, antibody-drug conjugates and degradation agents. The present review is the first to
cover a systemic and recent synopsis of tubulin-colchicine binding site agents. We believe that it would
provide an increase in our understanding of receptor-ligand interaction pattern and consciousness of a
series of challenges about tubulin target druggability.
Collapse
Affiliation(s)
- Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Liang Tian
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yanna Mao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Chuanjun Song
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
20
|
Shahein SA, Aboul-Enein AM, Higazy IM, Abou-Elella F, Lojkowski W, Ahmed ER, Mousa SA, AbouAitah K. Targeted anticancer potential against glioma cells of thymoquinone delivered by mesoporous silica core-shell nanoformulations with pH-dependent release. Int J Nanomedicine 2019; 14:5503-5526. [PMID: 31410001 PMCID: PMC6650459 DOI: 10.2147/ijn.s206899] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/10/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE Glioma is one of the most aggressive primary brain tumors and is incurable. Surgical resection, radiation, and chemotherapies have been the standard treatments for brain tumors, however, they damage healthy tissue. Therefore, there is a need for safe anticancer drug delivery systems. This is particularly true for natural prodrugs such as thymoquinone (TQ), which has a high therapeutic potential for cancers but has poor water solubility and insufficient targeting capacity. We have tailored novel core-shell nanoformulations for TQ delivery against glioma cells using mesoporous silica nanoparticles (MSNs) as a carrier. METHODS The core-shell nanoformulations were prepared with a core of MSNs loaded with TQ (MSNTQ), and the shell consisted of whey protein and gum Arabic (MSNTQ-WA), or chitosan and stearic acid (MSNTQ-CS). Nanoformulations were characterized, studied for release kinetics and evaluated for anticancer activity on brain cancer cells (SW1088 and A172) and cortical neuronal cells-2 (HCN2) as normal cells. Furthermore, they were evaluated for caspase-3, cytochrome c, cell cycle arrest, and apoptosis to understand the possible anticancer mechanism. RESULTS TQ release was pH-dependent and different for core and core-shell nanoformulations. A high TQ release from MSNTQ was detected at neutral pH 7.4, while a high TQ release from MSNTQ-WA and MSNTQ-CS was obtained at acidic pH 5.5 and 6.8, respectively; thus, TQ release in acidic tumor environment was enhanced. The release kinetics fitted with the Korsmeyer-Peppas kinetic model corresponding to diffusion-controlled release. Comparative in vitro tests with cancer and normal cells indicated a high anticancer efficiency for MSNTQ-WA compared to free TQ, and low cytotoxicity in the case of normal cells. The core-shell nanoformulations significantly improved caspase-3 activation, cytochrome c triggers, cell cycle arrest at G2/M, and apoptosis induction compared to TQ. CONCLUSION Use of MSNs loaded with TQ permit improved cancer targeting and opens the door to translating TQ into clinical application. Particularly good results were obtained for MSNTQ-WA.
Collapse
Affiliation(s)
- Samar A Shahein
- Biochemistry Department, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Ahmed M Aboul-Enein
- Biochemistry Department, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Iman M Higazy
- Department of Pharmaceutical Technology, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), Giza, Egypt
| | - Faten Abou-Elella
- Biochemistry Department, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Witold Lojkowski
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Esam R Ahmed
- Confirmatory Diagnostic Unit, Egyptian Organization for Vaccine, Sera and Biological Products (VACSERA), Giza, Egypt
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, New York, NY, USA
| | - Khaled AbouAitah
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
- Medicinal and Aromatic Plants Research Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), Giza, Egypt
| |
Collapse
|
21
|
Yang J, Li Y, Yan W, Li W, Qiu Q, Ye H, Chen L. Covalent modification of Cys-239 in β-tubulin by small molecules as a strategy to promote tubulin heterodimer degradation. J Biol Chem 2019; 294:8161-8170. [PMID: 30940730 DOI: 10.1074/jbc.ra118.006325] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/23/2019] [Indexed: 02/05/2023] Open
Abstract
Clinical microtubule-targeting drugs are functionally divided into microtubule-destabilizing and microtubule-stabilizing agents. Drugs from both classes achieve microtubule inhibition by binding different sites on tubulin and inhibiting or promoting polymerization with no concomitant effects on the protein levels of tubulin heterodimers. Here, we have identified a series of small molecules with diverse structures potentially representing a third class of novel tubulin inhibitors that promote degradation by covalent binding to Cys-239 of β-tubulin. The small molecules highlighted in this study include T0070907 (a peroxisome proliferator-activated receptor γ inhibitor), T007-1 (a T0070907 derivative), T138067, N,N'-ethylene-bis(iodoacetamide) (EBI), and allyl isothiocyanate (AITC). Label-free quantitative proteomic analysis revealed that T007-1 promotes tubulin degradation with high selectivity. Mass spectrometry findings showed covalent binding of both T0070907 and T007-01 to Cys-239 of β-tubulin. Furthermore, T007-1 exerted a degradative effect on tubulin isoforms possessing Cys-239 (β2, β4, and β5(β)) but not those containing Ser-239 (β3, β6) or mutant β-tubulin with a C239S substitution. Three small molecules (T138067, EBI, and AITC) also reported to bind covalently to Cys-239 of β-tubulin similarly induced tubulin degradation. Our results strongly suggest that covalent modification of Cys-239 of β-tubulin by small molecules could serve as a novel strategy to promote tubulin heterodimer degradation. We propose that these small molecules represent a third novel class of tubulin inhibitor agents that exert their effects through degradation activity.
Collapse
Affiliation(s)
- Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weimin Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
22
|
Synthetic strigolactone analogues reveal anti-cancer activities on hepatocellular carcinoma cells. Bioorg Med Chem Lett 2018; 28:1077-1083. [PMID: 29456109 DOI: 10.1016/j.bmcl.2018.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma (HCC) remains one of the leading causes of death worldwide. The complex etiology is attributed to many factors like heredity, cirrhosis, hepatitis infections or the dysregulation of the different molecular pathways. Nevertheless, the current treatment regimens have either severe side effects or tumors gradually acquire resistance upon prolonged use. Thus, developing a new selective treatment for HCC is the need of the hour. Many anticancer agents derived from plants have been evaluated for their cytotoxicity towards many human cancer cell lines. Strigolactones (SLs)-a newly discovered class of phytohormones, play a crucial role in the development of plant-root and shoot. Recently, many synthetic analogues of SLs have demonstrated pro-apoptotic effects on different cancer cell lines like prostate, breast, colon and lung. In this study, we tested synthetic SLs analogues on HCC cell line-HepG2 and evaluated their capability to induce cell proliferation inhibition and apoptosis. Primary WST-1 assays, followed by annexin-V/7AAD staining, demonstrated the anti-proliferative effects. The SLs analogues TIT3 and TIT7 were found to significantly reduce HepG2 cell viability in a dose- and time-dependent manner and induce apoptosis. Interestingly, though TIT3 and TIT7 strongly affected cancer cell proliferation, both compounds showed moderate anti-proliferative effect on normal cells. Further, migration of cancer cells was suppressed upon treatment with TIT3 and TIT7 in a wound healing assay. In summary, these findings suggest that two SLs analogues TIT3 and TIT7 exert selective inhibitory effects on cancer cells most likely through targeting microtubules. SLs analogues could be used in future as potential anti-cancer candidates in chemotherapy.
Collapse
|
23
|
Chowdhury FA, Hossain MK, Mostofa AGM, Akbor MM, Bin Sayeed MS. Therapeutic Potential of Thymoquinone in Glioblastoma Treatment: Targeting Major Gliomagenesis Signaling Pathways. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4010629. [PMID: 29651429 PMCID: PMC5831880 DOI: 10.1155/2018/4010629] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 12/27/2017] [Indexed: 02/06/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the most devastating brain tumors with median survival of one year and presents unique challenges to therapy because of its aggressive behavior. Current treatment strategy involves surgery, radiotherapy, immunotherapy, and adjuvant chemotherapy even though optimal management requires a multidisciplinary approach and knowledge of potential complications from both the disease and its treatment. Thymoquinone (TQ), the main bioactive component of Nigella sativa L., has exhibited anticancer effects in numerous preclinical studies. Due to its multitargeting nature, TQ interferes in a wide range of tumorigenic processes and counteract carcinogenesis, malignant growth, invasion, migration, and angiogenesis. TQ can specifically sensitize tumor cells towards conventional cancer treatments and minimize therapy-associated toxic effects in normal cells. Its potential to enter brain via nasal pathway due to volatile nature of TQ adds another advantage in overcoming blood-brain barrier. In this review, we summarized the potential role of TQ in different signaling pathways in GBM that have undergone treatment with standard therapeutic modalities or with TQ. Altogether, we suggest further comprehensive evaluation of TQ in preclinical and clinical level to delineate its implied utility as novel therapeutics to combat the challenges for the treatment of GBM.
Collapse
Affiliation(s)
- Fabliha Ahmed Chowdhury
- Department of Clinical Pharmacy and Pharmacology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Md Kamal Hossain
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - A. G. M. Mostofa
- Department of Clinical Pharmacy and Pharmacology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Maruf Mohammad Akbor
- Department of Clinical Pharmacy and Pharmacology, University of Dhaka, Dhaka 1000, Bangladesh
| | | |
Collapse
|
24
|
Goyal SN, Prajapati CP, Gore PR, Patil CR, Mahajan UB, Sharma C, Talla SP, Ojha SK. Therapeutic Potential and Pharmaceutical Development of Thymoquinone: A Multitargeted Molecule of Natural Origin. Front Pharmacol 2017; 8:656. [PMID: 28983249 PMCID: PMC5613109 DOI: 10.3389/fphar.2017.00656] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 09/04/2017] [Indexed: 12/16/2022] Open
Abstract
Thymoquinone, a monoterpene molecule is chemically known as 2-methyl-5-isopropyl-1, 4-benzoquinone. It is abundantly present in seeds of Nigella sativa L. that is popularly known as black cumin or black seed and belongs to the family Ranunculaceae. A large number of studies have revealed that thymoquinone is the major active constituent in N. sativa oil this constituent is responsible for the majority of the pharmacological properties. The beneficial organoprotective activities of thymoquinone in experimental animal models of different human diseases are attributed to the potent anti-oxidant and anti-inflammatory properties. Thymoquinone has also been shown to alter numerous molecular and signaling pathways in many inflammatory and degenerative diseases including cancer. Thymoquinone has been reported to possess potent lipophilicity and limited bioavailability and exhibits light and heat sensitivity. Altogether, these physiochemical properties encumber the successful formulation for the delivery of drug in oral dosages form and restrict the pharmaceutical development. In recent past, many efforts were undertaken to improve the bioavailability for clinical usage by manipulating the physiochemical parameters. The present review aimed to provide insights regarding the physicochemical characteristics, pharmacokinetics and the methods to promote pharmaceutical development and endorse the clinical usage of TQ in future by overcoming the associated physiochemical obstacles. It also enumerates briefly the pharmacological and molecular targets of thymoquinone as well as the pharmacological properties in various diseases and the underlying molecular mechanism. Though, a convincing number of experimental studies are available but human studies are not available with thymoquinone despite of the long history of use of black cumin in different diseases. Thus, the clinical studies including pharmacokinetic studies and regulatory toxicity studies are required to encourage the clinical development of thymoquinone.
Collapse
Affiliation(s)
- Sameer N. Goyal
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
- SVKM Institute of PharmacyDhule, India
| | - Chaitali P. Prajapati
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Prashant R. Gore
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Chandragouda R. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Umesh B. Mahajan
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| | - Sandhya P. Talla
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, North Maharashtra UniversityShirpur, India
| | - Shreesh K. Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| |
Collapse
|
25
|
Asaduzzaman Khan M, Tania M, Fu S, Fu J. Thymoquinone, as an anticancer molecule: from basic research to clinical investigation. Oncotarget 2017; 8:51907-51919. [PMID: 28881699 PMCID: PMC5584300 DOI: 10.18632/oncotarget.17206] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023] Open
Abstract
Thymoquinone is an anticancer phytochemical commonly found in black cumin. In this review, we discuss the potential of thymoquinone as anticancer molecule, its mechanism of action and future usage in clinical applications. Thymoquinone exhibits anticancer activity via numerous mechanisms of action, specifically by showing selective antioxidant and oxidant activity, interfering with DNA structure, affecting carcinogenic signaling molecules/pathways and immunomodulation. In vitro activity of thymoquinone has been further implicated in animal models of cancer; however, no clinical application has been proven yet. This is the optimum time to focus on clinical trials for developing thymoquinone as a future drug in cancer therapeutics.
Collapse
Affiliation(s)
- Md Asaduzzaman Khan
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China.,Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Mousumi Tania
- Division of Computer Aided Drug Design, The Red-Green Computing Centre, Dhaka, Bangladesh
| | - Shangyi Fu
- The Honors College, University of Houston, Houston, Texas, United States
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
26
|
Elmaci I, Altinoz MA. Thymoquinone: An edible redox-active quinone for the pharmacotherapy of neurodegenerative conditions and glial brain tumors. A short review. Biomed Pharmacother 2016; 83:635-640. [DOI: 10.1016/j.biopha.2016.07.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/04/2016] [Accepted: 07/13/2016] [Indexed: 01/24/2023] Open
|
27
|
Gleizes C, Kreutter G, Abbas M, Kassem M, Constantinescu AA, Boisramé-Helms J, Yver B, Toti F, Kessler L. β cell membrane remodelling and procoagulant events occur in inflammation-driven insulin impairment: a GLP-1 receptor dependent and independent control. J Cell Mol Med 2015; 20:231-42. [PMID: 26607759 PMCID: PMC4727568 DOI: 10.1111/jcmm.12683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 08/14/2015] [Indexed: 01/11/2023] Open
Abstract
Inflammation and hyperglycaemia are associated with a prothrombotic state. Cell-derived microparticles (MPs) are the conveyors of active procoagulant tissue factor (TF) and circulate at high concentration in diabetic patients. Liraglutide, a glucagon-like peptide (GLP)-1 analogue, is known to promote insulin secretion and β-cell preservation. In this in vitro study, we examined the link between insulin impairment, procoagulant activity and plasma membrane remodelling, under inflammatory conditions. Rin-m5f β-cell function, TF activity mediated by MPs and their modulation by 1 μM liraglutide were examined in a cell cross-talk model. Methyl-β-cyclodextrine (MCD), a cholesterol depletor, was used to evaluate the involvement of raft on TF activity, MP shedding and insulin secretion as well as Soluble N-éthylmaleimide-sensitive-factor Attachment protein Receptor (SNARE)-dependent exocytosis. Cytokines induced a two-fold increase in TF activity at MP surface that was counteracted by liraglutide. Microparticles prompted TF activity on the target cells and a two-fold decrease in insulin secretion via protein kinase A (PKA) and p38 signalling, that was also abolished by liraglutide. Large lipid raft clusters were formed in response to cytokines and liraglutide or MCD-treated cells showed similar patterns. Cells pre-treated by saturating concentration of the GLP-1r antagonist exendin (9-39), showed a partial abolishment of the liraglutide-driven insulin secretion and liraglutide-decreased TF activity. Measurement of caspase 3 cleavage and MP shedding confirmed the contribution of GLP-1r-dependent and -independent pathways. Our results confirm an integrative β-cell response to GLP-1 that targets receptor-mediated signalling and membrane remodelling pointing at the coupling of insulin secretion and inflammation-driven procoagulant events.
Collapse
Affiliation(s)
- Céline Gleizes
- EA7293, Vascular and Tissular Stress in Transplantation, Faculty of Medicine, University of Strasbourg, Illkirch, France.,Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Guillaume Kreutter
- EA7293, Vascular and Tissular Stress in Transplantation, Faculty of Medicine, University of Strasbourg, Illkirch, France.,Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Malak Abbas
- EA7293, Vascular and Tissular Stress in Transplantation, Faculty of Medicine, University of Strasbourg, Illkirch, France.,Doctoral School of Sciences and Technologies, Lebanese University, Beiruth-Hadath, Lebanon
| | - Mohamad Kassem
- EA7293, Vascular and Tissular Stress in Transplantation, Faculty of Medicine, University of Strasbourg, Illkirch, France
| | - Andrei Alexandru Constantinescu
- EA7293, Vascular and Tissular Stress in Transplantation, Faculty of Medicine, University of Strasbourg, Illkirch, France.,Department of Parasitology and Parasitic Diseases and Animal Biology, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine, Bucharest, Romania
| | - Julie Boisramé-Helms
- Department of Reanimation, Nouvel hopital civil, Strasbourg CEDEX, France.,Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Blandine Yver
- EA7293, Vascular and Tissular Stress in Transplantation, Faculty of Medicine, University of Strasbourg, Illkirch, France
| | - Florence Toti
- UMR7213 CNRS, Laboratory of Biophotonics and Pharmacology, Faculty of Pharmacy, University of Strasbourg, Illkirch, France
| | - Laurence Kessler
- EA7293, Vascular and Tissular Stress in Transplantation, Faculty of Medicine, University of Strasbourg, Illkirch, France.,Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, Strasbourg, France.,Department of Diabetology, University Hospital, Strasbourg Cedex, France
| |
Collapse
|
28
|
Thymoquinone and its therapeutic potentials. Pharmacol Res 2015; 95-96:138-58. [DOI: 10.1016/j.phrs.2015.03.011] [Citation(s) in RCA: 251] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/16/2015] [Accepted: 03/16/2015] [Indexed: 12/11/2022]
|
29
|
Engelmann D, Meier C, Alla V, Pützer BM. A balancing act: orchestrating amino-truncated and full-length p73 variants as decisive factors in cancer progression. Oncogene 2014; 34:4287-99. [PMID: 25381823 DOI: 10.1038/onc.2014.365] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/24/2014] [Accepted: 09/29/2014] [Indexed: 12/23/2022]
Abstract
p73 is the older sibling of p53 and mimics most of its tumor-suppressor functions. Through alternative promoter usage and splicing, the TP73 gene generates more than two dozen isoforms of which N-terminal truncated DNp73 variants have a decisive role in cancer pathogenesis as they outweigh the positive effects of full-length TAp73 and p53 in acting as a barrier to tumor development. Beyond the prevailing view that DNp73 predominantly counteract cell cycle arrest and apoptosis, latest progress indicates that these isoforms acquire novel functions in epithelial-to-mesenchymal transition, metastasis and therapy resistance. New insight into the mechanisms underlying this behavior reinforced the expectation that DNp73 variants contribute to aggressive cellular traits through both loss of wild-type tumor-suppressor activity and gain-of-function, suggesting an equally important role in cancer progression as mutant p53. In this review, we describe the novel properties of DNp73 in the invasion metastasis cascade and outline the comprehensive p73 regulatome with an emphasis on molecular processes putting TAp73 out of action in advanced tumors. These intriguing insights provoke a new understanding of the acquisition of aggressive traits by cancer cells and may help to set novel therapies for a broad range of metastatic tumors.
Collapse
Affiliation(s)
- D Engelmann
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
| | - C Meier
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
| | - V Alla
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
| | - B M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
30
|
Antitumor activity of monoterpenes found in essential oils. ScientificWorldJournal 2014; 2014:953451. [PMID: 25401162 PMCID: PMC4220615 DOI: 10.1155/2014/953451] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 12/15/2022] Open
Abstract
Cancer is a complex genetic disease that is a major public health problem worldwide, accounting for about 7 million deaths each year. Many anticancer drugs currently used clinically have been isolated from plant species or are based on such substances. Accumulating data has revealed anticancer activity in plant-derived monoterpenes. In this review the antitumor activity of 37 monoterpenes found in essential oils is discussed. Chemical structures, experimental models, and mechanisms of action for bioactive substances are presented.
Collapse
|
31
|
Kundu J, Chun KS, Aruoma OI, Kundu JK. Mechanistic perspectives on cancer chemoprevention/chemotherapeutic effects of thymoquinone. Mutat Res 2014; 768:22-34. [PMID: 25847385 DOI: 10.1016/j.mrfmmm.2014.05.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 05/20/2014] [Indexed: 01/16/2023]
Abstract
The bioactive natural products (plant secondary metabolites) are widely known to possess therapeutic value for the prevention and treatment of various chronic diseases including cancer. Thymoquinone (2-methyl-5-isopropyl-1,4-benzoquinone; TQ), a monoterpene present in black cumin seeds, exhibits pleiotropic pharmacological activities including antioxidant, anti-inflammatory, antidiabetic and antitumor effects. TQ inhibits experimental carcinogenesis in a wide range of animal models and has been shown to arrest the growth of various cancer cells in culture as well as xenograft tumors in vivo. The mechanistic basis of anticancer effects of TQ includes the inhibition of carcinogen metabolizing enzyme activity and oxidative damage of cellular macromolecules, attenuation of inflammation, induction of cell cycle arrest and apoptosis in tumor cells, blockade of tumor angiogenesis, and suppression of migration, invasion and metastasis of cancer cells. TQ shows synergistic and/or potentiating anticancer effects when combined with clinically used chemotherapeutic agents. At the molecular level, TQ targets various components of intracellular signaling pathways, particularly a variety of upstream kinases and transcription factors, which are aberrantly activated during the course of tumorigenesis.
Collapse
Affiliation(s)
- Juthika Kundu
- College of Pharmacy, Keimyung University, Daegu 704 701, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 704 701, Republic of Korea
| | - Okezie I Aruoma
- School of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA.
| | - Joydeb Kumar Kundu
- College of Pharmacy, Keimyung University, Daegu 704 701, Republic of Korea.
| |
Collapse
|
32
|
Gleizes C, Constantinescu A, Abbas M, Bouhadja H, Zobairi F, Kessler L, Toti F. Liraglutide protects Rin-m5f β cells by reducing procoagulant tissue factor activity and apoptosis prompted by microparticles under conditions mimicking Instant Blood-Mediated Inflammatory Reaction. Transpl Int 2014; 27:733-40. [DOI: 10.1111/tri.12286] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/02/2013] [Accepted: 02/14/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Céline Gleizes
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
| | - Andrei Constantinescu
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
- Department of Parasitology and Parasitic Diseases and Animal Biology; Faculty of Veterinary Medicine; University of Agronomical Sciences and Veterinary Medicine; Bucharest Romania
| | - Malak Abbas
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
- Faculty of Sciences; Lebanon University; Hadath Lebanon
| | - Houda Bouhadja
- Faculty of Pharmacy; University of Strasbourg; UMR7213 CNRS; Laboratory of Biophotonics and Pharmacology; Illkirch France
| | - Fatiha Zobairi
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
| | - Laurence Kessler
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
- Department of Diabetology; University Hospital; Strasbourg Cedex France
| | - Florence Toti
- Faculty of Pharmacy; University of Strasbourg; UMR7213 CNRS; Laboratory of Biophotonics and Pharmacology; Illkirch France
| |
Collapse
|
33
|
Acharya BR, Chatterjee A, Ganguli A, Bhattacharya S, Chakrabarti G. Thymoquinone inhibits microtubule polymerization by tubulin binding and causes mitotic arrest following apoptosis in A549 cells. Biochimie 2014; 97:78-91. [DOI: 10.1016/j.biochi.2013.09.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/26/2013] [Indexed: 10/26/2022]
|
34
|
Constantinescu AA, Gleizes C, Alhosin M, Yala E, Zobairi F, Leclercq A, Stoian G, Mitrea IL, Prévost G, Toti F, Kessler L. Exocrine cell-derived microparticles in response to lipopolysaccharide promote endocrine dysfunction in cystic fibrosis. J Cyst Fibros 2013; 13:219-26. [PMID: 24095207 DOI: 10.1016/j.jcf.2013.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 08/26/2013] [Accepted: 08/30/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Diabetes in cystic fibrosis (CF) is a result of exocrine pancreas alteration followed by endocrine dysfunction at a later stage. Microparticles (MPs) are plasma membrane fragments shed from stimulated or damaged cells that act as cellular effectors. Our aim was to identify a new form of interaction between exocrine and endocrine pancreatic cells mediated by exocrine MPs, in the context of recurrent infection in CF. METHODS MPs from either human exocrine CFTRΔF508-mutated (CFPAC-1) cells or exocrine normal pancreatic (PANC-1) cells were collected after treatment by LPS from Pseudomonas aeruginosa and applied to rat endocrine normal insulin-secreting RIN-m5F cells. MP membrane integration in target cells was established by confocal microscopy and flow cytometry using PKH26 lipid probe. Apoptosis, lysosomal activity, insulin secretion were measured after 18 h. MP-mediated NF-κB activation was measured in HEK-Blue reporter cells by SEAP reporter gene system and in RIN-m5F cells by Western blot. In endocrine normal cells, CFTR inhibition was achieved using Inhibitor-172. RESULTS Compared to PANC-1, MPs from CFPAC-1 significantly reduced insulin secretion and lysosomal activity in RIN-m5F. MPs induced NF-κB activation by increasing the level of IκB phosphorylation. Moreover, the inhibition of NF-κB activation using specific inhibitors was associated with a restored insulin secretion. Interestingly, CFTR inhibition in normal RIN-m5F cells promoted apoptosis and decreased insulin secretion. CONCLUSIONS During recurrent infections associated with CF, exocrine MPs may contribute to endocrine cell dysfunction via NF-κB pathways. Membrane CFTR dysfunction is associated with decreased insulin secretion.
Collapse
Affiliation(s)
- Andrei Alexandru Constantinescu
- EA7293, Vascular and Tissular Stress in Transplantation, Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, 74 route du Rhin, F-67401 Illkirch, Strasbourg, France; Department of Parasitology and Parasitic Diseases and Animal Biology, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine, 105 spl. Independentei, sector 5, 050097 Bucharest, Romania
| | - Céline Gleizes
- EA7293, Vascular and Tissular Stress in Transplantation, Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, 74 route du Rhin, F-67401 Illkirch, Strasbourg, France
| | - Mahmoud Alhosin
- UMR7213 CNRS, Laboratory of Biophotonics and Pharmacology, 74 route du Rhin, F-67401 Illkirch, France
| | - Elhassan Yala
- EA7293, Vascular and Tissular Stress in Transplantation, Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, 74 route du Rhin, F-67401 Illkirch, Strasbourg, France
| | - Fatiha Zobairi
- EA7293, Vascular and Tissular Stress in Transplantation, Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, 74 route du Rhin, F-67401 Illkirch, Strasbourg, France
| | - Alexandre Leclercq
- Department of Pneumology, University Hospital, 1 place de L'hôpital, CHU de Strasbourg, BP426, 67091 Strasbourg Cedex, France
| | - Gheorghe Stoian
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 spl. Independentei, sector 5, 050095 Bucharest, Romania
| | - Ioan Liviu Mitrea
- Department of Parasitology and Parasitic Diseases and Animal Biology, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine, 105 spl. Independentei, sector 5, 050097 Bucharest, Romania
| | - Gilles Prévost
- EA7290 Early Bacterial Virulence, Faculty of Medicine, University of Strasbourg, 3 rue Koeberlé, F-67000 Strasbourg, France
| | - Florence Toti
- UMR7213 CNRS, Laboratory of Biophotonics and Pharmacology, 74 route du Rhin, F-67401 Illkirch, France; Faculty of Pharmacy, University of Strasbourg, 74 route du Rhin, F-67401 Illkirch, France
| | - Laurence Kessler
- EA7293, Vascular and Tissular Stress in Transplantation, Federation of Translational Medicine of Strasbourg, Faculty of Medicine, University of Strasbourg, 74 route du Rhin, F-67401 Illkirch, Strasbourg, France; Department of Diabetology, University Hospital, 1 place de l'Hôpital, CHU de Strasbourg, BP421, 67091 Strasbourg Cedex, France.
| |
Collapse
|
35
|
Racoma IO, Meisen WH, Wang QE, Kaur B, Wani AA. Thymoquinone inhibits autophagy and induces cathepsin-mediated, caspase-independent cell death in glioblastoma cells. PLoS One 2013; 8:e72882. [PMID: 24039814 PMCID: PMC3767730 DOI: 10.1371/journal.pone.0072882] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 07/16/2013] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma is the most aggressive and common type of malignant brain tumor in humans, with a median survival of 15 months. There is a great need for more therapies for the treatment of glioblastoma. Naturally occurring phytochemicals have received much scientific attention because many exhibit potent tumor killing action. Thymoquinone (TQ) is the bioactive compound of the Nigella sativa seed oil. TQ has anti-oxidant, anti-inflammatory and anti-neoplastic actions with selective cytotoxicity for human cancer cells compared to normal cells. Here, we show that TQ selectively inhibits the clonogenicity of glioblastoma cells as compared to normal human astrocytes. Also, glioblastoma cell proliferation could be impaired by chloroquine, an autophagy inhibitor, suggesting that glioblastoma cells may be dependent on the autophagic pathway for survival. Exposure to TQ caused an increase in the recruitment and accumulation of the microtubule-associated protein light chain 3-II (LC3-II). TQ also caused an accumulation of the LC3-associated protein p62, confirming the inhibition of autophagy. Furthermore, the levels of Beclin-1 protein expression were unchanged, indicating that TQ interferes with a later stage of autophagy. Finally, treatment with TQ induces lysosome membrane permeabilization, as determined by a specific loss of red acridine orange staining. Lysosome membrane permeabilization resulted in a leakage of cathepsin B into the cytosol, which mediates caspase-independent cell death that can be prevented by pre-treatment with a cathepsin B inhibitor. TQ induced apoptosis, as determined by an increase in PI and Annexin V positive cells. However, apoptosis appears to be caspase-independent due to failure of the caspase inhibitor z-VAD-FMK to prevent cell death and absence of the typical apoptosis related signature DNA fragmentation. Inhibition of autophagy is an exciting and emerging strategy in cancer therapy. In this vein, our results describe a novel mechanism of action for TQ as an autophagy inhibitor selectively targeting glioblastoma cells.
Collapse
Affiliation(s)
- Ira O. Racoma
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Walter Hans Meisen
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Qi-En Wang
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Balveen Kaur
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Altaf A. Wani
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
36
|
Schneider-Stock R, Fakhoury IH, Zaki AM, El-Baba CO, Gali-Muhtasib HU. Thymoquinone: fifty years of success in the battle against cancer models. Drug Discov Today 2013; 19:18-30. [PMID: 24001594 DOI: 10.1016/j.drudis.2013.08.021] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/04/2013] [Accepted: 08/23/2013] [Indexed: 12/15/2022]
Abstract
Thymoquinone (TQ), the main active constituent of black seed essential oil, exhibits promising effects against inflammatory diseases and cancer. TQ, modulates signaling pathways that are key to cancer progression, and enhances the anticancer potential of clinical drugs while reducing their toxic side effects. Considering that TQ was isolated 50 years ago, this review focuses on TQ's chemical and pharmacological properties and the latest advances in TQ analog design and nanoformulation. We discuss our current state of knowledge of TQ's adjuvant potential and in vivo antitumor activity and highlight its ability to modulate the hallmarks of cancer.
Collapse
Affiliation(s)
- Regine Schneider-Stock
- Experimental Tumor Pathology, Institute for Pathology, University of Erlangen-Nuremberg, Germany
| | | | - Angela M Zaki
- Department of Biology, American University of Beirut, Lebanon
| | - Chirine O El-Baba
- Experimental Tumor Pathology, Institute for Pathology, University of Erlangen-Nuremberg, Germany
| | | |
Collapse
|
37
|
Soldevilla B, Millán CS, Bonilla F, Domínguez G. The TP73 complex network: ready for clinical translation in cancer? Genes Chromosomes Cancer 2013; 52:989-1006. [PMID: 23913810 DOI: 10.1002/gcc.22095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/28/2013] [Indexed: 01/05/2023] Open
Abstract
TP73 is a member of the TP53 family, whose deregulated expression has been reported in a wide variety of cancers and linked to patients' outcome. The fact that TP73 encodes a complex number of isoforms (TAp73 and ΔTAp73) with opposing functions and the cross-talk with other members of the family (TP53 and TP63) make it difficult to determine its clinical relevance. Here, we review the molecular mechanisms driving TAp73 and ΔTAp73 expression and how these variants inhibit or promote carcinogenesis. We also highlight the intricate interplay between TP53 family members. In addition, we comment on current pharmacological approaches targeting the TP73 pathway and those affecting the TAp73/ΔTAp73 ratio. Finally, we discuss the current data available in the literature that provide evidence on the role of TP73 variants in predicting prognosis. To date, most of the studies that evaluate the status levels of TP73 isoforms have been based on limited-size series. Despite this limitation, these publications highlight the correlation between high levels of the oncogenic forms and failure to respond to chemotherapy and/or shorter survival. Finally, we emphasize the need for studies to evaluate the significance of combining the deregulation of various members of the TP53 family in order to define patient outcome or their responsiveness to specific therapies.
Collapse
Affiliation(s)
- Beatriz Soldevilla
- Servicio de Oncología Médica, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | | | | | | |
Collapse
|