1
|
Ji Y, Lin Y, He J, Xie Y, An W, Luo X, Qiao X, Li Z. Research progress of mitochondria and cytoskeleton crosstalk in tumour development. Biochim Biophys Acta Rev Cancer 2025; 1880:189254. [PMID: 39732178 DOI: 10.1016/j.bbcan.2024.189254] [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: 07/11/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
During tumour progression, organelle function undergoes dramatic changes, and crosstalk among organelles plays a significant role. Crosstalk between mitochondria and other organelles such as the endoplasmic reticulum and cytoskeleton has focussed attention on the mechanisms of tumourigenesis. This review demonstrates an overview of the molecular structure of the mitochondrial-cytoskeletal junction and its biological interactions. It also presents a detailed and comprehensive description of mitochondrial-cytoskeletal crosstalk in tumour occurrence and development, including tumour cell proliferation, apoptosis, autophagy, metabolic rearrangement, and metastasis. Finally, the application of crosstalk in tumour therapy, including drug combinations and chemoresistance, is discussed. This review offers a theoretical basis for establishing mitochondrial-cytoskeletal junctions as therapeutic targets, and offers novel insights into the future management of malignant tumours.
Collapse
Affiliation(s)
- Yue Ji
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China
| | - Yingchi Lin
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China; Provincial key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, China
| | - Jing He
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Diseases, Shenyang 110002, Liaoning Province, China
| | - Yuanyuan Xie
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China
| | - Wenmin An
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China
| | - Xinyu Luo
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China
| | - Xue Qiao
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China; Department of Central Laboratory, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China.
| | - Zhenning Li
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China.
| |
Collapse
|
2
|
Lin RJ, Xie L, Gao TY, Yang YZ, Huang L, Cheng K, Chen ZP. Design, synthesis and anti-tumor evaluation of novel pyrimidine and quinazoline analogues. Eur J Med Chem 2025; 282:117057. [PMID: 39561497 DOI: 10.1016/j.ejmech.2024.117057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/29/2024] [Accepted: 11/12/2024] [Indexed: 11/21/2024]
Abstract
Disrupting microtubule dynamics has emerged as a promising strategy for cancer therapy. Novel trimethoxyanilino-substituted pyrimidine and quinazoline derivatives were designed and synthesized to serve as potent microtubule-inhibiting agents with anti-proliferative activity. Compound 2k demonstrates high efficacy against B16-F10 cancer cells at low nanomolar concentrations, with an IC50 of 0.098 ± 0.006 μM, which is comparable to colchicine. Mechanistic studies have revealed that 2k has the ability to inhibit microtubule protein polymerization in vitro, resulting in cell cycle arrest and apoptosis. Furthermore, 2k inhibits tumor cell migration and exhibits significant anti-tumor efficacy in a melanoma tumor model without causing obvious toxicity. In summary, the pyrimidine derivative 2k exhibits excellent anticancer activity and provides a new scaffold for the development of novel microtubule inhibitors, which deserves further in-depth research.
Collapse
Affiliation(s)
- Ren-Jie Lin
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lin Xie
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Yu Gao
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yi-Zhou Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lan Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Zhi-Peng Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Merlin JPJ, Crous A, Abrahamse H. Combining Photodynamic Therapy and Targeted Drug Delivery Systems: Enhancing Mitochondrial Toxicity for Improved Cancer Outcomes. Int J Mol Sci 2024; 25:10796. [PMID: 39409125 PMCID: PMC11477455 DOI: 10.3390/ijms251910796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer treatment continues to be a substantial problem due to tumor complexities and persistence, demanding novel therapeutic techniques. This review investigates the synergistic potential of combining photodynamic therapy (PDT) and tailored medication delivery technologies to increase mitochondrial toxicity and improve cancer outcomes. PDT induces selective cellular damage and death by activating photosensitizers (PS) with certain wavelengths of light. However, PDT's efficacy can be hampered by issues such as poor light penetration and a lack of selectivity. To overcome these challenges, targeted drug delivery systems have emerged as a promising technique for precisely delivering therapeutic medicines to tumor cells while avoiding off-target effects. We investigate how these technologies can improve mitochondrial targeting and damage, which is critical for causing cancer cell death. The combination method seeks to capitalize on the advantages of both modalities: selective PDT activation and specific targeted drug delivery. We review current preclinical and clinical evidence supporting the efficacy of this combination therapy, focusing on case studies and experimental models. This review also addresses issues such as safety, distribution efficiency, resistance mechanisms, and costs. The prospects of further research include advances in photodynamic agents and medication delivery technology, with a focus on personalized treatment. In conclusion, combining PDT with targeted drug delivery systems provides a promising frontier in cancer therapy, with the ability to overcome current treatment limits and open the way for more effective, personalized cancer treatments.
Collapse
Affiliation(s)
- J. P. Jose Merlin
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa; (A.C.); (H.A.)
| | | | | |
Collapse
|
4
|
Wu Q, Wang Y, Liu J, Guan X, Chang X, Liu Z, Liu R. Microtubules and cardiovascular diseases: insights into pathology and therapeutic strategies. Int J Biochem Cell Biol 2024; 175:106650. [PMID: 39237031 DOI: 10.1016/j.biocel.2024.106650] [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: 03/21/2024] [Revised: 08/25/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Microtubules, complex cytoskeletal structures composed of tubulin proteins in eukaryotic cells, have garnered recent attention in cardiovascular research. Investigations have focused on the post-translational modifications of tubulin, including acetylation and detyrosination. Perturbations in microtubule homeostasis have been implicated in various pathological processes associated with cardiovascular diseases such as heart failure, ischemic heart disease, and arrhythmias. Thus, elucidating the intricate interplay between microtubule dynamics and cardiovascular pathophysiology is imperative for advancing preventive and therapeutic strategies. Several natural compounds have been identified to potentially modulate microtubules, thereby exerting regulatory effects on cardiovascular diseases. This review synthesizes current literature to delineate the roles of microtubules in cardiovascular diseases and assesses the potential of natural compounds in microtubule-targeted therapies.
Collapse
Affiliation(s)
- Qiaomin Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yanli Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jinfeng Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xuanke Guan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Zhiming Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ruxiu Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
5
|
Jiang F, Yu M, Liang Y, Ding K, Wang Y. Discovery of Novel Diaryl-Substituted Fused Heterocycles Targeting Katanin and Tubulin with Potent Antitumor and Antimultidrug Resistance Efficacy. J Med Chem 2024; 67:12118-12142. [PMID: 38996194 DOI: 10.1021/acs.jmedchem.4c00878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Disrupting microtubule dynamics has emerged as a promising strategy for cancer treatment. However, drug resistance remains a challenge hindering the development of microtubule-targeting agents. In this work, a novel class of diaryl substituted fused heterocycles were designed, synthesized, and evaluated, which were demonstrated as effective dual katanin and tubulin regulators with antitumor activity. Following three rounds of stepwise optimization, compound 21b, featuring a 3H-imidazo[4,5-b]pyridine core, displayed excellent targeting capabilities on katanin and tubulin, along with notable antiproliferative and antimetastatic effects. Mechanistic studies revealed that 21b disrupts the microtubule network in tumor cells, leading to G2/M cell cycle arrest and apoptosis induction. Importantly, 21b exhibited significant inhibition of tumor growth in MDA-MB-231 and A549/T xenograft tumor models without evident toxicity and side effects. In conclusion, compound 21b presents a novel mechanism for disrupting microtubule dynamics, warranting further investigation as a dual-targeted antitumor agent with potential antimultidrug resistance properties.
Collapse
Affiliation(s)
- Fuhao Jiang
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Min Yu
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yuru Liang
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Kuiling Ding
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yang Wang
- School of Pharmacy, Fudan University, Shanghai 201203, China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
6
|
Singh D. A sojourn on mitochondria targeted drug delivery systems for cancer: Strategies, clinical and future prospects. Mitochondrion 2024; 74:101826. [PMID: 38092248 DOI: 10.1016/j.mito.2023.101826] [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: 10/10/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Mitochondria, often referred to as the powerhouses of the cell, have emerged as promising targets for cancer therapy due to their pivotal roles in cell survival, apoptosis, and energy metabolism. This sojourn emphasizes the significance of mitochondria-targeted drug delivery systems in cancer therapeutics. The unique characteristics of cancer cell mitochondria, such as altered membrane potential and distinct lipid composition, offer an avenue for selective drug targeting. Several strategies have been explored to exploit these features, including the use of lipophilic cations, mitochondria-penetrating peptides, and nanocarriers tailored for mitochondrial delivery. Mitochondria-targeted drug delivery systems have demonstrated enhanced therapeutic efficacy and reduced systemic toxicity in preclinical models. Some of these systems have made a successful transition to clinical trials, illustrating their potential in real-world oncology settings. However, there remain challenges like intracellular barriers, potential off-target effects, and the complexity of tumor heterogeneity that must be addressed to fully harness the potential of mitochondria-targeted drug delivery systems. As research progresses, it is anticipated that innovative approaches and technologies will be developed to improve the specificity and efficacy of mitochondrial targeting, paving the way for more effective and safer cancer treatments in the future. This review serves as a comprehensive guide to the current state of mitochondria-targeted drug delivery systems for cancer, highlighting key strategies, clinical progress, and prospective avenues for future research.
Collapse
Affiliation(s)
- Dilpreet Singh
- University Institute of Pharmaceutical Sciences, Chandigarh University, Mohali, India.
| |
Collapse
|
7
|
Kozubek M, Denner TC, Eckert M, Hoenke S, Csuk R. On the influence of the rhodamine substituents onto the cytotoxicity of mitocanic maslinic acid rhodamine conjugates. RESULTS IN CHEMISTRY 2023. [DOI: 10.1016/j.rechem.2022.100708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
8
|
Zunica ERM, Axelrod CL, Kirwan JP. Phytochemical Targeting of Mitochondria for Breast Cancer Chemoprevention, Therapy, and Sensitization. Int J Mol Sci 2022; 23:ijms232214152. [PMID: 36430632 PMCID: PMC9692881 DOI: 10.3390/ijms232214152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
Breast cancer is a common and deadly disease that causes tremendous physical, emotional, and financial burden on patients and society. Early-stage breast cancer and less aggressive subtypes have promising prognosis for patients, but in aggressive subtypes, and as cancers progress, treatment options and responses diminish, dramatically decreasing survival. Plants are nutritionally rich and biologically diverse organisms containing thousands of metabolites, some of which have chemopreventive, therapeutic, and sensitizing properties, providing a rich source for drug discovery. In this study we review the current landscape of breast cancer with a central focus on the potential role of phytochemicals for treatment, management, and disease prevention. We discuss the relevance of phytochemical targeting of mitochondria for improved anti-breast cancer efficacy. We highlight current applications of phytochemicals and derivative structures that display anti-cancer properties and modulate cancer mitochondria, while describing future applicability and identifying areas of promise.
Collapse
|
9
|
Romagnoli R, Oliva P, Prencipe F, Manfredini S, Budassi F, Brancale A, Ferla S, Hamel E, Corallo D, Aveic S, Manfreda L, Mariotto E, Bortolozzi R, Viola G. Design, Synthesis and Biological Investigation of 2-Anilino Triazolopyrimidines as Tubulin Polymerization Inhibitors with Anticancer Activities. Pharmaceuticals (Basel) 2022; 15:1031. [PMID: 36015179 PMCID: PMC9415608 DOI: 10.3390/ph15081031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
A further investigation aiming to generate new potential antitumor agents led us to synthesize a new series of twenty-two compounds characterized by the presence of the 7-(3',4',5'-trimethoxyphenyl)-[1,2,4]triazolo[1,5-a]pyrimidine pharmacophore modified at its 2-position. Among the synthesized compounds, three were significantly more active than the others. These bore the substituents p-toluidino (3d), p-ethylanilino (3h) and 3',4'-dimethylanilino (3f), and these compounds had IC50 values of 30-43, 160-240 and 67-160 nM, respectively, on HeLa, A549 and HT-29 cancer cells. The p-toluidino derivative 3d was the most potent inhibitor of tubulin polymerization (IC50: 0.45 µM) and strongly inhibited the binding of colchicine to tubulin (72% inhibition), with antiproliferative activity superior to CA-4 against A549 and HeLa cancer cell lines. In vitro investigation showed that compound 3d was able to block treated cells in the G2/M phase of the cell cycle and to induce apoptosis following the intrinsic pathway, further confirmed by mitochondrial depolarization and caspase-9 activation. In vivo experiments conducted on the zebrafish model showed good activity of 3d in reducing the mass of a HeLa cell xenograft. These effects occurred at nontoxic concentrations to the animal, indicating that 3d merits further developmental studies.
Collapse
Affiliation(s)
- Romeo Romagnoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paola Oliva
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Filippo Prencipe
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
| | - Stefano Manfredini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Federica Budassi
- Medicinal Chemistry Department, Integrated Drug Discovery, Aptuit-An Evotec Company, 37135 Verona, Italy
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Salvatore Ferla
- Faculty of Medicine, Health and Life Science, Swansea University Medical School, Grove Building, Swansea University, Swansea SA2 8PP, UK
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Lorenzo Manfreda
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Elena Mariotto
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Roberta Bortolozzi
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Giampietro Viola
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| |
Collapse
|
10
|
Tian C, Wang M, Shi X, Chen X, Wang X, Zhang Z, Liu J. Discovery of (2-(pyrrolidin-1-yl)thieno[3,2-d]pyrimidin-4-yl)(3,4,5-trimethoxyphenyl)methanone as a novel potent tubulin depolymerizing and vascular disrupting agent. Eur J Med Chem 2022; 238:114466. [DOI: 10.1016/j.ejmech.2022.114466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/03/2022] [Accepted: 05/12/2022] [Indexed: 11/15/2022]
|
11
|
Liu GY, Chen S, Lee G, Shaiv K, Chen P, Cheng H, Hong S, Yang W, Huang S, Chang Y, Wang H, Kao C, Sun P, Chao M, Lee Y, Tang M, Lin Y. Precise control of microtubule disassembly in living cells. EMBO J 2022; 41:e110472. [PMID: 35686621 PMCID: PMC9340485 DOI: 10.15252/embj.2021110472] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/15/2022] [Accepted: 05/05/2022] [Indexed: 12/28/2022] Open
Abstract
Microtubules tightly regulate various cellular activities. Our understanding of microtubules is largely based on experiments using microtubule-targeting agents, which, however, are insufficient to dissect the dynamic mechanisms of specific microtubule populations, due to their slow effects on the entire pool of microtubules. To overcome this technological limitation, we have used chemo and optogenetics to disassemble specific microtubule subtypes, including tyrosinated microtubules, primary cilia, mitotic spindles, and intercellular bridges, by rapidly recruiting engineered microtubule-cleaving enzymes onto target microtubules in a reversible manner. Using this approach, we show that acute microtubule disassembly swiftly halts vesicular trafficking and lysosomal dynamics. It also immediately triggers Golgi and ER reorganization and slows the fusion/fission of mitochondria without affecting mitochondrial membrane potential. In addition, cell rigidity is increased after microtubule disruption owing to increased contractile stress fibers. Microtubule disruption furthermore prevents cell division, but does not cause cell death during interphase. Overall, the reported tools facilitate detailed analysis of how microtubules precisely regulate cellular architecture and functions.
Collapse
Affiliation(s)
- Grace Y Liu
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shiau‐Chi Chen
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Gang‐Hui Lee
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan
| | - Kritika Shaiv
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Pin‐Yu Chen
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Hsuan Cheng
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shi‐Rong Hong
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Wen‐Ting Yang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shih‐Han Huang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ya‐Chu Chang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Hsien‐Chu Wang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ching‐Lin Kao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Pin‐Chiao Sun
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Hong Chao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Yian‐Ying Lee
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Jer Tang
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan
| | - Yu‐Chun Lin
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
- Department of Medical ScienceNational Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
12
|
Oliva P, Romagnoli R, Cacciari B, Manfredini S, Padroni C, Brancale A, Ferla S, Hamel E, Corallo D, Aveic S, Milan N, Mariotto E, Viola G, Bortolozzi R. Synthesis and Biological Evaluation of Highly Active 7-Anilino Triazolopyrimidines as Potent Antimicrotubule Agents. Pharmaceutics 2022; 14:1191. [PMID: 35745764 PMCID: PMC9230136 DOI: 10.3390/pharmaceutics14061191] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/22/2022] [Accepted: 05/31/2022] [Indexed: 12/14/2022] Open
Abstract
Two different series of fifty-two compounds, based on 3',4',5'-trimethoxyaniline (7a-ad) and variably substituted anilines (8a-v) at the 7-position of the 2-substituted-[1,2,4]triazolo [1,5-a]pyrimidine nucleus, had moderate to potent antiproliferative activity against A549, MDA-MB-231, HeLa, HT-29 and Jurkat cancer cell lines. All derivatives with a common 3-phenylpropylamino moiety at the 2-position of the triazolopyrimidine scaffold and different halogen-substituted anilines at its 7-position, corresponding to 4'-fluoroaniline (8q), 4'-fluoro-3'-chloroaniline (8r), 4'-chloroaniline (8s) and 4'-bromoaniline (8u), displayed the greatest antiproliferative activity with mean IC50's of 83, 101, 91 and 83 nM, respectively. These four compounds inhibited tubulin polymerization about 2-fold more potently than combretastatin A-4 (CA-4), and their activities as inhibitors of [3H]colchicine binding to tubulin were similar to that of CA-4. These data underlined that the 3',4',5'-trimethoxyanilino moiety at the 7-position of the [1,2,4]triazolo [1,5-a]pyrimidine system, which characterized compounds 7a-ad, was not essential for maintaining potent antiproliferative and antitubulin activities. Compounds 8q and 8r had high selectivity against cancer cells, and their interaction with tubulin led to the accumulation of HeLa cells in the G2/M phase of the cell cycle and to apoptotic cell death through the mitochondrial pathway. Finally, compound 8q significantly inhibited HeLa cell growth in zebrafish embryos.
Collapse
Affiliation(s)
- Paola Oliva
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (P.O.); (B.C.)
| | - Romeo Romagnoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (P.O.); (B.C.)
| | - Barbara Cacciari
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (P.O.); (B.C.)
| | - Stefano Manfredini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy;
| | - Chiara Padroni
- Medicinal Chemistry Department, Integrated Drug Discovery, Aptuit—An Evotec Company, Via A. Fleming, 37135 Verona, Italy;
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK;
| | - Salvatore Ferla
- Swansea University Medical School, Institute of Life Sciences 2, Swansea University, Swansea SA2 8PP, UK;
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA;
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy; (D.C.); (S.A.)
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy; (D.C.); (S.A.)
| | - Noemi Milan
- Hemato-Oncology Lab, Department of Woman’s and Child’s Health, University of Padova, 35131 Padova, Italy; (N.M.); (E.M.); (R.B.)
| | - Elena Mariotto
- Hemato-Oncology Lab, Department of Woman’s and Child’s Health, University of Padova, 35131 Padova, Italy; (N.M.); (E.M.); (R.B.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy
| | - Giampietro Viola
- Hemato-Oncology Lab, Department of Woman’s and Child’s Health, University of Padova, 35131 Padova, Italy; (N.M.); (E.M.); (R.B.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy
| | - Roberta Bortolozzi
- Hemato-Oncology Lab, Department of Woman’s and Child’s Health, University of Padova, 35131 Padova, Italy; (N.M.); (E.M.); (R.B.)
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Corso Stati Uniti 4, 35128 Padova, Italy
| |
Collapse
|
13
|
βIII-Tubulin Structural Domains Regulate Mitochondrial Network Architecture in an Isotype-Specific Manner. Cells 2022; 11:cells11050776. [PMID: 35269398 PMCID: PMC8909761 DOI: 10.3390/cells11050776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/12/2022] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
βIII-tubulin is a neuronal microtubule protein that is aberrantly expressed in epithelial cancers. The microtubule network is implicated in regulating the architecture and dynamics of the mitochondrial network, although the isotype-specific role for β-tubulin proteins that constitute this microtubule network remains unclear. High-resolution electron microscopy revealed that manipulation of βIII-tubulin expression levels impacts the volume and shape of mitochondria. Analysis of the structural domains of the protein identifies that the C-terminal tail of βIII-tubulin, which distinguishes this protein from other β-tubulin isotypes, significantly contributes to the isotype-specific effects of βIII-tubulin on mitochondrial architecture. Mass spectrometry analysis of protein–protein interactions with β-tubulin isotypes identifies that βIII-tubulin specifically interacts with regulators of mitochondrial dynamics that may mediate these functional effects. Advanced quantitative dynamic lattice light sheet imaging of the mitochondrial network reveals that βIII-tubulin promotes a more dynamic and extended reticular mitochondrial network, and regulates mitochondrial volume. A regulatory role for the βIII-tubulin C-terminal tail in mitochondrial network dynamics and architecture has widespread implications for the maintenance of mitochondrial homeostasis in health and disease.
Collapse
|
14
|
van der Westhuizen D, Bezuidenhout DI, Munro OQ. Cancer molecular biology and strategies for the design of cytotoxic gold(I) and gold(III) complexes: a tutorial review. Dalton Trans 2021; 50:17413-17437. [PMID: 34693422 DOI: 10.1039/d1dt02783b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This tutorial review highlights key principles underpinning the design of selected metallodrugs to target specific biological macromolecules (DNA and proteins). The review commences with a descriptive overview of the eukaryotic cell cycle and the molecular biology of cancer, particularly apoptosis, which is provided as a necessary foundation for the discovery, design, and targeting of metal-based anticancer agents. Drugs which target DNA have been highlighted and clinically approved metallodrugs discussed. A brief history of the development of mainly gold-based metallodrugs is presented prior to addressing ligand systems for stabilizing and adding functionality to bio-active gold(I) and gold(III) complexes, particularly in the burgeoning field of anticancer metallodrugs. Concepts such as multi-modal and selective cytotoxic agents are covered where necessary for selected compounds. The emerging role of carbenes as the ligand system of choice to achieve these goals for gold-based metallodrug candidates is highlighted prior to closing the review with comments on some future directions that this research field might follow. The latter section ultimately emphasizes the importance of understanding the fate of metal complexes in cells to garner key mechanistic insights.
Collapse
Affiliation(s)
- Danielle van der Westhuizen
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg 2050, South Africa.
| | - Daniela I Bezuidenhout
- Laboratory of Inorganic Chemistry, Environmental and Chemical Engineering, University of Oulu, P. O. Box 3000, 90014 Oulu, Finland.
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg 2050, South Africa.
| |
Collapse
|
15
|
Rostovtseva TK, Bezrukov SM, Hoogerheide DP. Regulation of Mitochondrial Respiration by VDAC Is Enhanced by Membrane-Bound Inhibitors with Disordered Polyanionic C-Terminal Domains. Int J Mol Sci 2021; 22:7358. [PMID: 34298976 PMCID: PMC8306229 DOI: 10.3390/ijms22147358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/18/2022] Open
Abstract
The voltage-dependent anion channel (VDAC) is the primary regulating pathway of water-soluble metabolites and ions across the mitochondrial outer membrane. When reconstituted into lipid membranes, VDAC responds to sufficiently large transmembrane potentials by transitioning to gated states in which ATP/ADP flux is reduced and calcium flux is increased. Two otherwise unrelated cytosolic proteins, tubulin, and α-synuclein (αSyn), dock with VDAC by a novel mechanism in which the transmembrane potential draws their disordered, polyanionic C-terminal domains into and through the VDAC channel, thus physically blocking the pore. For both tubulin and αSyn, the blocked state is observed at much lower transmembrane potentials than VDAC gated states, such that in the presence of these cytosolic docking proteins, VDAC's sensitivity to transmembrane potential is dramatically increased. Remarkably, the features of the VDAC gated states relevant for bioenergetics-reduced metabolite flux and increased calcium flux-are preserved in the blocked state induced by either docking protein. The ability of tubulin and αSyn to modulate mitochondrial potential and ATP production in vivo is now supported by many studies. The common physical origin of the interactions of both tubulin and αSyn with VDAC leads to a general model of a VDAC inhibitor, facilitates predictions of the effect of post-translational modifications of known inhibitors, and points the way toward the development of novel therapeutics targeting VDAC.
Collapse
Affiliation(s)
- Tatiana K. Rostovtseva
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Sergey M. Bezrukov
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
| | - David P. Hoogerheide
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA;
| |
Collapse
|
16
|
Mercier AE, Prudent R, Pepper MS, De Koning L, Nolte E, Peronne L, Nel M, Lafanechère L, Joubert AM. Characterization of Signalling Pathways That Link Apoptosis and Autophagy to Cell Death Induced by Estrone Analogues Which Reversibly Depolymerize Microtubules. Molecules 2021; 26:molecules26030706. [PMID: 33572896 PMCID: PMC7866274 DOI: 10.3390/molecules26030706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022] Open
Abstract
The search for novel anti-cancer compounds which can circumvent chemotherapeutic drug resistance and limit systemic toxicity remains a priority. 2-Ethyl-3-O-sulphamoyl-estra-1,3,5(10)15-tetraene-3-ol-17one (ESE-15-one) and 2-ethyl-3-O-sulphamoyl-estra-1,3,5(10)16-tetraene (ESE-16) are sulphamoylated 2-methoxyestradiol (2-ME) analogues designed by our research team. Although their cytotoxicity has been demonstrated in vitro, the temporal and mechanistic responses of the initiated intracellular events are yet to be determined. In order to do so, assays investigating the compounds' effects on microtubules, cell cycle progression, signalling cascades, autophagy and apoptosis were conducted using HeLa cervical- and MDA-MB-231 metastatic breast cancer cells. Both compounds reversibly disrupted microtubule dynamics as an early event by binding to the microtubule colchicine site, which blocked progression through the cell cycle at the G1/S- and G2/M transitions. This was supported by increased pRB and p27Kip1 phosphorylation. Induction of apoptosis with time-dependent signalling involving the p-JNK, Erk1/2 and Akt/mTOR pathways and loss of mitochondrial membrane potential was demonstrated. Inhibition of autophagy attenuated the apoptotic response. In conclusion, the 2-ME analogues induced a time-dependent cross-talk between cell cycle checkpoints, apoptotic signalling and autophagic processes, with an increased reactive oxygen species formation and perturbated microtubule functioning appearing to connect the processes. Subtle differences in the responses were observed between the two compounds and the different cell lines.
Collapse
Affiliation(s)
- Anne E. Mercier
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (E.N.); (M.N.); (L.L.); (A.M.J.)
- Correspondence: ; Tel.: +27-(0)-12-319-2141
| | - Renaud Prudent
- Institute for Advanced Biosciences, Team Regulation and Pharmacology of the Cytoskeleton, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38700 Grenoble, France; (R.P.); (L.P.)
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, School of Medicine, SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Leanne De Koning
- RPPA Platform, Institut Curie Centre de Recherche, PSL Research University, Paris 75248, France;
| | - Elsie Nolte
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (E.N.); (M.N.); (L.L.); (A.M.J.)
| | - Lauralie Peronne
- Institute for Advanced Biosciences, Team Regulation and Pharmacology of the Cytoskeleton, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38700 Grenoble, France; (R.P.); (L.P.)
| | - Marcel Nel
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (E.N.); (M.N.); (L.L.); (A.M.J.)
| | - Laurence Lafanechère
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (E.N.); (M.N.); (L.L.); (A.M.J.)
- Institute for Advanced Biosciences, Team Regulation and Pharmacology of the Cytoskeleton, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38700 Grenoble, France; (R.P.); (L.P.)
| | - Anna M. Joubert
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (E.N.); (M.N.); (L.L.); (A.M.J.)
| |
Collapse
|
17
|
Romagnoli R, Oliva P, Salvador MK, Manfredini S, Padroni C, Brancale A, Ferla S, Hamel E, Ronca R, Maccarinelli F, Rruga F, Mariotto E, Viola G, Bortolozzi R. A facile synthesis of diaryl pyrroles led to the discovery of potent colchicine site antimitotic agents. Eur J Med Chem 2021; 214:113229. [PMID: 33550186 DOI: 10.1016/j.ejmech.2021.113229] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 12/13/2022]
Abstract
Three different series of cis-restricted analogues of combretastatin A-4 (CA-4), corresponding to thirty-nine molecules that contained a pyrrole nucleus interposed between the two aryl rings, were prepared by a palladium-mediated coupling approach and evaluated for their antiproliferative activity against six human cancer cell lines. In the two series of 1,2-diaryl pyrrole derivatives, results suggested that the presence of the 3',4',5'-trimethoxyphenyl moiety at the N-1 position of the pyrrole ring was more favorable for antiproliferative activity. In the series of 3,4-diarylpyrrole analogues, three compounds (11i-k) exhibited maximal antiproliferative activity, showing excellent antiproliferative activity against the CA-4 resistant HT-29 cells. Inhibition of tubulin polymerization of selected 1,2 pyrrole derivatives (9a, 9c, 9o and 10a) was similar to that observed with CA-4, while the isomeric 3,4-pyrrole analogues 11i-k were generally from 1.5- to 2-fold more active than CA-4. Compounds 11j and 11k were the only compounds that showed activity as inhibitors of colchicine binding comparable to that CA-4. Compound 11j had biological properties consistent with its intracellular target being tubulin. This compound was able to block the cell cycle in metaphase and to induce significant apoptosis at a concentration of 25 nM, following the mitochondrial pathway, with low toxicity for normal cells. More importantly, compound 11j exerted activity in vivo superior to that of CA-4P, being able to significantly reduce tumor growth in a syngeneic murine tumor model even at the lower dose tested (5.0 mg/kg).
Collapse
Affiliation(s)
- Romeo Romagnoli
- Dipartimento di Scienze Chimiche, Farmaceutiche e Agrarie, Via Luigi Borsari 46, Università Degli Studi di Ferrara, 44121, Ferrara, Italy.
| | - Paola Oliva
- Dipartimento di Scienze Chimiche, Farmaceutiche e Agrarie, Via Luigi Borsari 46, Università Degli Studi di Ferrara, 44121, Ferrara, Italy
| | - Maria Kimatrai Salvador
- Dipartimento di Scienze Chimiche, Farmaceutiche e Agrarie, Via Luigi Borsari 46, Università Degli Studi di Ferrara, 44121, Ferrara, Italy
| | - Stefano Manfredini
- Dipartimento di Scienze Della Vita e Biotecnologie, Università Degli Studi di Ferrara, 44121, Ferrara, Italy
| | - Chiara Padroni
- Medicinal Chemistry Department, Integrated Drug Discovery, Aptuit, An Evotec Company, Via A. Fleming 4, 37135, Verona, Italy
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | | | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Roberto Ronca
- Dipartimento di Medicina Molecolare e Traslazionale Unità di Oncologia Sperimentale Ed Immunologia, Università di Brescia, 25123, Brescia, Italy
| | - Federica Maccarinelli
- Dipartimento di Medicina Molecolare e Traslazionale Unità di Oncologia Sperimentale Ed Immunologia, Università di Brescia, 25123, Brescia, Italy
| | - Fatlum Rruga
- Dipartimento di Salute Della Donna e Del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131, Padova, Italy
| | - Elena Mariotto
- Dipartimento di Salute Della Donna e Del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131, Padova, Italy
| | - Giampietro Viola
- Dipartimento di Salute Della Donna e Del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131, Padova, Italy; Istituto di Ricerca Pediatrica (IRP), Corso Stati Uniti 4, 35128, Padova, Italy.
| | - Roberta Bortolozzi
- Istituto di Ricerca Pediatrica (IRP), Corso Stati Uniti 4, 35128, Padova, Italy.
| |
Collapse
|
18
|
Spanò V, Barreca M, Rocca R, Bortolozzi R, Bai R, Carbone A, Raimondi MV, Piccionello AP, Montalbano A, Alcaro S, Hamel E, Viola G, Barraja P. Insight on [1,3]thiazolo[4,5-e]isoindoles as tubulin polymerization inhibitors. Eur J Med Chem 2020; 212:113122. [PMID: 33401199 DOI: 10.1016/j.ejmech.2020.113122] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
A series of [1,3]thiazolo[4,5-e]isoindoles has been synthesized through a versatile and high yielding multistep sequence. Evaluation of the antiproliferative activity of the new compounds on the full NCI human tumor cell line panel highlighted several compounds that are able to inhibit tumor cell proliferation at micromolar-submicromolar concentrations. The most active derivative 11g was found to cause cell cycle arrest at the G2/M phase and induce apoptosis in HeLa cells, following the mitochondrial pathway, making it a lead compound for the discovery of new antimitotic drugs.
Collapse
Affiliation(s)
- Virginia Spanò
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Marilia Barreca
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Roberta Rocca
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Græcia di Catanzaro, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Academic Spinoff, Università Magna Græcia di Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Roberta Bortolozzi
- Istituto di Ricerca Pediatrica IRP, Fondazione Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, United States
| | - Anna Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Maria Valeria Raimondi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Antonio Palumbo Piccionello
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Alessandra Montalbano
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy.
| | - Stefano Alcaro
- Net4Science srl, Academic Spinoff, Università Magna Græcia di Catanzaro, Viale Europa, 88100, Catanzaro, Italy; Dipartimento di Scienze della Salute, Università Magna Græcia di Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, United States
| | - Giampietro Viola
- Istituto di Ricerca Pediatrica IRP, Fondazione Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy; Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, via Giustiniani 2, 35131, Padova, Italy.
| | - Paola Barraja
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| |
Collapse
|
19
|
Spanò V, Rocca R, Barreca M, Giallombardo D, Montalbano A, Carbone A, Raimondi MV, Gaudio E, Bortolozzi R, Bai R, Tassone P, Alcaro S, Hamel E, Viola G, Bertoni F, Barraja P. Pyrrolo[2',3':3,4]cyclohepta[1,2- d][1,2]oxazoles, a New Class of Antimitotic Agents Active against Multiple Malignant Cell Types. J Med Chem 2020; 63:12023-12042. [PMID: 32986419 PMCID: PMC7901646 DOI: 10.1021/acs.jmedchem.0c01315] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Indexed: 02/08/2023]
Abstract
A new class of pyrrolo[2',3':3,4]cyclohepta[1,2-d][1,2]oxazoles was synthesized for the treatment of hyperproliferative pathologies, including neoplasms. The new compounds were screened in the 60 human cancer cell lines of the NCI drug screen and showed potent activity with GI50 values reaching the nanomolar level, with mean graph midpoints of 0.08-0.41 μM. All compounds were further tested on six lymphoma cell lines, and eight showed potent growth inhibitory effects with IC50 values lower than 500 nM. Mechanism of action studies showed the ability of the new [1,2]oxazoles to arrest cells in the G2/M phase in a concentration dependent manner and to induce apoptosis through the mitochondrial pathway. The most active compounds inhibited tubulin polymerization, with IC50 values of 1.9-8.2 μM, and appeared to bind to the colchicine site. The G2/M arrest was accompanied by apoptosis, mitochondrial depolarization, generation of reactive oxygen species, and PARP cleavage.
Collapse
Affiliation(s)
- Virginia Spanò
- Department of Biological, Chemical and Pharmaceutical
Sciences and Technologies (STEBICEF), University of Palermo,
Via Archirafi 32, 90123 Palermo, Italy
| | - Roberta Rocca
- Net4Science srl, Academic Spinoff,
Università Magna Græcia di Catanzaro, Viale Europa, 88100
Catanzaro, Italy
- Dipartimento di Medicina Sperimentale e Clinica,
Università Magna Græcia di Catanzaro, Viale
Europa, 88100 Catanzaro, Italy
| | - Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical
Sciences and Technologies (STEBICEF), University of Palermo,
Via Archirafi 32, 90123 Palermo, Italy
- Institute of Oncology Research, Faculty
of Biomedical Sciences, Università della Svizzera Italiana, Via
Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Daniele Giallombardo
- Department of Biological, Chemical and Pharmaceutical
Sciences and Technologies (STEBICEF), University of Palermo,
Via Archirafi 32, 90123 Palermo, Italy
| | - Alessandra Montalbano
- Department of Biological, Chemical and Pharmaceutical
Sciences and Technologies (STEBICEF), University of Palermo,
Via Archirafi 32, 90123 Palermo, Italy
| | - Anna Carbone
- Department of Biological, Chemical and Pharmaceutical
Sciences and Technologies (STEBICEF), University of Palermo,
Via Archirafi 32, 90123 Palermo, Italy
| | - Maria Valeria Raimondi
- Department of Biological, Chemical and Pharmaceutical
Sciences and Technologies (STEBICEF), University of Palermo,
Via Archirafi 32, 90123 Palermo, Italy
| | - Eugenio Gaudio
- Institute of Oncology Research, Faculty
of Biomedical Sciences, Università della Svizzera Italiana, Via
Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Roberta Bortolozzi
- Istituto di Ricerca Pediatrica IRP,
Fondazione Città della Speranza, Corso Stati Uniti 4, 35127 Padova,
Italy
| | - Ruoli Bai
- Screening Technologies Branch, Developmental Therapeutics
Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for
Cancer Research, National Cancer Institute, National Institutes of
Health, Frederick, Maryland 21702, United States
| | - Pierfrancesco Tassone
- Dipartimento di Medicina Sperimentale e Clinica,
Università Magna Græcia di Catanzaro, Viale
Europa, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute,
Università Magna Græcia di Catanzaro, Viale
Europa, 88100 Catanzaro, Italy
- Net4Science srl, Academic Spinoff,
Università Magna Græcia di Catanzaro, Viale Europa, 88100
Catanzaro, Italy
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics
Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for
Cancer Research, National Cancer Institute, National Institutes of
Health, Frederick, Maryland 21702, United States
| | - Giampietro Viola
- Istituto di Ricerca Pediatrica IRP,
Fondazione Città della Speranza, Corso Stati Uniti 4, 35127 Padova,
Italy
- Dipartimento di Salute della Donna e del Bambino,
Laboratorio di Oncoematologia, Università di Padova, Via
Giustiniani 2, 35131 Padova, Italy
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty
of Biomedical Sciences, Università della Svizzera Italiana, Via
Vincenzo Vela 6, 6500 Bellinzona, Switzerland
- Oncology Institute of Southern
Switzerland, Via Ospedale, 6500 Bellinzona,
Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical
Sciences and Technologies (STEBICEF), University of Palermo,
Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
20
|
Kwon A, Lee GB, Park T, Lee JH, Ko P, You E, Ahn JH, Eom SH, Rhee S, Song WK. Potent Small-Molecule Inhibitors Targeting Acetylated Microtubules as Anticancer Agents Against Triple-Negative Breast Cancer. Biomedicines 2020; 8:E338. [PMID: 32917017 PMCID: PMC7555225 DOI: 10.3390/biomedicines8090338] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Microtubules are one of the major targets for anticancer drugs because of their role in cell proliferation and migration. However, as anticancer drugs targeting microtubules have side effects, including the death of normal cells, it is necessary to develop anticancer agents that can target microtubules by specifically acting on cancer cells only. In this study, we identified chemicals that can act as anticancer agents by specifically binding to acetylated microtubules, which are predominant in triple-negative breast cancer (TNBC). The chemical compounds disrupted acetylated microtubule lattices by interfering with microtubule access to alpha-tubulin acetyltransferase 1 (αTAT1), a major acetyltransferase of microtubules, resulting in the increased apoptotic cell death of MDA-MB-231 cells (a TNBC cell line) compared with other cells, such as MCF-10A and MCF-7, which lack microtubule acetylation. Moreover, mouse xenograft experiments showed that treatment with the chemical compounds markedly reduced tumor growth progression. Taken together, the newly identified chemical compounds can be selective for acetylated microtubules and act as potential therapeutic agents against microtubule acetylation enrichment in TNBC.
Collapse
Affiliation(s)
- Ahreum Kwon
- Cell Logistics and Silver Health Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Gwi Bin Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (G.B.L.); (T.P.); (J.H.A.); (S.H.E.)
| | - Taein Park
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (G.B.L.); (T.P.); (J.H.A.); (S.H.E.)
| | - Jung Hoon Lee
- Department of Biochemistry and Cell Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA;
| | - Panseon Ko
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea; (P.K.); (E.Y.)
| | - Eunae You
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea; (P.K.); (E.Y.)
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (G.B.L.); (T.P.); (J.H.A.); (S.H.E.)
| | - Soo Hyun Eom
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (G.B.L.); (T.P.); (J.H.A.); (S.H.E.)
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea; (P.K.); (E.Y.)
| | - Woo Keun Song
- Cell Logistics and Silver Health Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| |
Collapse
|
21
|
Sugita Y, Takao K, Uesawa Y, Nagai J, Iijima Y, Sano M, Sakagami H. Development of Newly Synthesized Chromone Derivatives with High Tumor Specificity against Human Oral Squamous Cell Carcinoma. MEDICINES (BASEL, SWITZERLAND) 2020; 7:E50. [PMID: 32858984 PMCID: PMC7555025 DOI: 10.3390/medicines7090050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/23/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022]
Abstract
Since many anticancer drugs show severe adverse effects such as mucositis, peripheral neurotoxicity, and extravasation, it was crucial to explore new compounds with much reduced adverse effects. Comprehensive investigation with human malignant and nonmalignant cells demonstrated that derivatives of chromone, back-bone structure of flavonoid, showed much higher tumor specificity as compared with three major polyphenols in the natural kingdom, such as lignin-carbohydrate complex, tannin, and flavonoid. A total 291 newly synthesized compounds of 17 groups (consisting of 12 chromones, 2 esters, and 3 amides) gave a wide range of the intensity of tumor specificity, possibly reflecting the fitness for the optimal 3D structure and electric state. Among them, 7-methoxy-3-[(1E)-2-phenylethenyl]-4H-1-benzopyran-4-one (compound 22), which belongs to 3-styrylchromones, showed the highest tumor specificity. 22 induced subG1 and G2 + M cell population in human oral squamous cell carcinoma cell line, with much less keratinocyte toxicity as compared with doxorubicin and 5-FU. However, 12 active compounds selected did not necessarily induce apoptosis and mitotic arrest. This compound can be used as a lead compound to manufacture more active compound.
Collapse
Affiliation(s)
- Yoshiaki Sugita
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Koichi Takao
- Department of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama 350-0295, Japan
| | - Yoshihiro Uesawa
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Tokyo 204-858, Japan
| | - Junko Nagai
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Tokyo 204-858, Japan
| | - Yosuke Iijima
- Department of Oral and Maxillofacial Surgery, Saitama Medical Center, Saitama Medical University, Kawagoe 350-8550, Japan
| | - Motohiko Sano
- Division of Applied Pharmaceutical Education and Research, Hoshi University, Tokyo 142-8501, Japan
| | - Hiroshi Sakagami
- Meikai University Research Institute of Odontology (M-RIO), 1-1 Keyakidai, Sakado, Saitama 350-0283, Japan
| |
Collapse
|
22
|
Zhu H, Sun H, Liu Y, Duan Y, Liu J, Yang X, Li W, Qin S, Xu S, Zhu Z, Xu J. Design, synthesis and biological evaluation of vinyl selenone derivatives as novel microtubule polymerization inhibitors. Eur J Med Chem 2020; 207:112716. [PMID: 32853870 DOI: 10.1016/j.ejmech.2020.112716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/06/2020] [Accepted: 07/22/2020] [Indexed: 01/16/2023]
Abstract
A series of novel vinyl selenone derivatives were designed, synthesized and evaluated as the tubulin polymerization inhibitors using a bioisosteric strategy. Among them, the representative compound 11k exhibited satisfactory anti-proliferative activities with IC50 values ranging from 0.287 to 0.621 μM against a panel of cancer cell lines. Importantly, 11k displayed more potent in vivo antitumor activity than the positive control paclitaxel, CA-4 and parent compound 4 without apparent toxicity, which was presumably ascribed to the antiangiogenic, antiproliferative and selective effects of selenium, along with the unique physiological activity of indole skeleton, which were both introduced into the structure of target compounds. Further mechanism study demonstrated that compound 11k showed potent activity in tubulin polymerization inhibition with IC50 value of 1.82 μM. Moreover, cellular mechanism studies disclosed that 11k blocked cell cycle arrest at G2/M phase, induced cell apoptosis and depolarized mitochondria of K562 cells. Meanwhile, 11k reduced the cell migration and had potent vascular disrupting activity. In summary, 11k could serve as a promising lead for the development of more efficient microtubule polymerization inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Huajian Zhu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Honghao Sun
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Yang Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Yiping Duan
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China; Department of Organic Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Jie Liu
- Department of Organic Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Xue Yang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China; Department of Organic Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Wenlong Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Shuai Qin
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China.
| |
Collapse
|
23
|
Design, synthesis, in vitro and in vivo biological evaluation of 2-amino-3-aroylbenzo[b]furan derivatives as highly potent tubulin polymerization inhibitors. Eur J Med Chem 2020; 200:112448. [DOI: 10.1016/j.ejmech.2020.112448] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/09/2020] [Accepted: 05/09/2020] [Indexed: 01/02/2023]
|
24
|
Shuai W, Li X, Li W, Xu F, Lu L, Yao H, Yang L, Zhu H, Xu S, Zhu Z, Xu J. Design, synthesis and anticancer properties of isocombretapyridines as potent colchicine binding site inhibitors. Eur J Med Chem 2020; 197:112308. [DOI: 10.1016/j.ejmech.2020.112308] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
|
25
|
NMK-BH2, a novel microtubule-depolymerising bis (indolyl)-hydrazide-hydrazone, induces apoptotic and autophagic cell death in cervical cancer cells by binding to tubulin at colchicine - site. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118762. [PMID: 32502617 DOI: 10.1016/j.bbamcr.2020.118762] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/10/2020] [Accepted: 05/27/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Microtubules, the key components of the eukaryotic cytoskeleton and mitotic spindle, are one of the most sought-after targets for cancer chemotherapy, especially due to their indispensible role in mitosis. Cervical cancer is a prevalent malignancy among women of developing countries including India. In spite of the remarkable therapeutic advancement, the non-specificity of chemotherapeutic drugs adversely affect the patients' survival and well-being, thus, necessitating the quest for novel indole-based anti-microtubule agent against cervical cancer, with high degree of potency and selectivity. METHODS For in vitro studies, we used MTT assay, confocal microscopy, fluorescence microscopy, flow cytometry and Western blot analysis. Study in cell free system was accomplished by spectrophotometry, fluorescence spectroscopy and TEM and computational analysis was done by AutodockTools 1.5.6. RESULTS NMK-BH2 exhibited significant and selective anti-proliferative activity against cervical cancer HeLa cells (IC50 = 1.5 μM) over normal cells. It perturbed the cytoskeletal and spindle microtubules of HeLa cells leading to mitotic block and cell death by apoptosis and autophagy. Furthermore, NMK-BH2 targeted the tubulin-microtubule system through fast and strong binding to the αβ-tubulin heterodimers at colchicine-site. CONCLUSION This study identifies and characterises NMK-BH2 as a novel anti-microtubule agent and provides insights into its key anti-cancer mechanism through two different cell death pathways: apoptosis and autophagy, which are mutually independent. GENERAL SIGNIFICANCE It navigates the potential of the novel bis (indolyl)-hydrazide-hydrazone, NMK-BH2, to serve as lead for development of new generation microtubule-disrupting chemotherapeutic with improved efficacy and remarkable selectivity towards better cure of cervical cancer.
Collapse
|
26
|
Chakraborty S, Dlie ZY, Mukherjee B, Besra SE, Sengupta S, Sen R, Mukherjee A. A Comparative Investigation of the Ability of Various Aptamer-Functionalized Drug Nanocarriers to Induce Selective Apoptosis in Neoplastic Hepatocytes: In Vitro and In Vivo Outcome. AAPS PharmSciTech 2020; 21:89. [PMID: 32026264 DOI: 10.1208/s12249-020-1629-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
Aptamers offer a significant promise to target various cancers including hepatocellular carcinoma (HCC), for their high affinity and ability to reach the target site(s), non-immunogenicity, and low cost. The targeting ability to neoplastic hepatocytes by the aptamer, TLS 9a with phosphorothioate backbone modification (designated as L5), has not been explored yet. Hence, we investigated the comparative potential of L5 with some other previously reported liver cancer cell-specific aptamers, conjugated on the surface of drug-nanocarriers. Various in vitro studies such as cytotoxicity, in vitro cellular uptake, cell cycle analysis, and investigations related to apoptosis were performed. In vivo studies carried out here include macroscopic and microscopic hepatic alterations in chemically induced hepatocarcinogenesis in rats, upon experimental treatments. The outcome of the investigations revealed that L5-functionalized drug-nanocarrier (PTX-NPL5) had the highest apoptotic potential compared with the other aptamer-conjugated experimental formulations. Further, its maximum internalization by neoplastic hepatocytes and minimum internalization by normal hepatocytes indicate that it had the potential to preferentially target the neoplastic hepatocytes. Data of in vivo studies revealed that PTX-NPL5 reduced tumor incidences and tumor progress. Superior potency of PTX-NPL5 may be due to the maximum affinity of L5 towards neoplastic hepatocytes resulting in maximum permeation of drug-nanocarrier in them. An effective site-specific targeting of neoplastic hepatocytes can be achieved by L5 for preferential delivery of therapeutics. Further, investigations are needed to identify the target protein(s) on neoplastic hepatocytes responsible for ligand-receptor interaction of L5.
Collapse
|
27
|
Chakraborty S, Dlie ZY, Chakraborty S, Roy S, Mukherjee B, Besra SE, Dewanjee S, Mukherjee A, Ojha PK, Kumar V, Sen R. Aptamer-Functionalized Drug Nanocarrier Improves Hepatocellular Carcinoma toward Normal by Targeting Neoplastic Hepatocytes. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:34-49. [PMID: 32146417 PMCID: PMC7063179 DOI: 10.1016/j.omtn.2020.01.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 12/14/2022]
Abstract
Site-specific delivery of chemotherapeutics specifically to neoplastic hepatocytes without affecting normal hepatocytes should be a focus for potential therapeutic management of hepatocellular carcinoma (HCC). The aptamer TLS 9a with phosphorothioate backbone modifications (L5) has not been explored so far for preferential delivery of therapeutics in neoplastic hepatocytes to induce apoptosis. Thus, the objective of the present investigation was to compare the therapeutic potential of L5-functionalized drug nanocarrier (PTX-NPL5) with those of the other experimental drug nanocarriers functionalized by previously reported HCC cell-targeting aptamers and non-aptamer ligands, such as galactosamine and apotransferrin. A myriad of well-defined investigations such as cell cycle analysis, TUNEL (terminal deoxynucleotidyltransferase-mediated deoxyuridine triphosphate nick end labeling) assay, and studies related to apoptosis, histopathology, and immunoblotting substantiated that PTX-NPL5 had the highest potency among the different ligand-attached experimental formulations in inducing selective apoptosis in neoplastic hepatocytes via a mitochondrial-dependent apoptotic pathway. PTX-NPL5 did not produce any notable toxic effects in healthy hepatocytes, thus unveiling a new and a safer option in targeted therapy for HCC. Molecular modeling study identified two cell-surface biomarker proteins (tumor-associated glycoprotein 72 [TAG-72] and heat shock protein 70 [HSP70]) responsible for ligand-receptor interaction of L5 and preferential internalization of PTX-NPL5 via clathrin-mediated endocytosis in neoplastic hepatocytes. The potential of PTX-NPL5 has provided enough impetus for its rapid translation from the pre-clinical to clinical domain to establish itself as a targeted therapeutic to significantly prolong survival in HCC.
Collapse
Affiliation(s)
- Samrat Chakraborty
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Zewdu Yilma Dlie
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Somdyuti Chakraborty
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Somdatta Roy
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Biswajit Mukherjee
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India.
| | - Shila Elizabeth Besra
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Alankar Mukherjee
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Probir Kumar Ojha
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Vinay Kumar
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Ramkrishna Sen
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India; Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
28
|
Wang LJ, Chiou JT, Lee YC, Huang CH, Shi YJ, Chang LS. SIRT3, PP2A and TTP protein stability in the presence of TNF-α on vincristine-induced apoptosis of leukaemia cells. J Cell Mol Med 2020; 24:2552-2565. [PMID: 31930676 PMCID: PMC7028858 DOI: 10.1111/jcmm.14949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/27/2019] [Accepted: 12/16/2019] [Indexed: 01/15/2023] Open
Abstract
The contribution of vincristine (VCR)-induced microtubule destabilization to evoke apoptosis in cancer cells remains to be resolved. Thus, we investigated the cytotoxic mechanism of VCR on U937 and HL-60 human leukaemia cell lines. We discovered that VCR treatment resulted in the up-regulation of TNF-α expression and activation of the death receptor pathway, which evoked apoptosis of U937 cells. Moreover, VCR induced microtubule destabilization and mitotic arrest. VCR treatment down-regulated SIRT3, and such down-regulation caused mitochondrial ROS to initiate phosphorylation of p38 MAPK. p38 MAPK suppressed MID1-modulated degradation of the protein phosphatase 2A (PP2A) catalytic subunit. The SIRT3-ROS-p38 MAPK-PP2A axis inhibited tristetraprolin (TTP)-controlled TNF-α mRNA degradation, consequently, up-regulating TNF-α expression. Restoration of SIRT3 and TTP expression, or inhibition of the ROS-p38 MAPK axis increased the survival of VCR-treated cells and repressed TNF-α up-regulation. In contrast to suppression of the ROS-p38 MAPK axis, overexpression of SIRT3 modestly inhibited the effect of VCR on microtubule destabilization and mitotic arrest in U937 cells. Apoptosis of HL-60 cells, similarly, went through the same pathway. Collectively, our data indicate that the SIRT3-ROS-p38 MAPK-PP2A-TTP axis modulates TNF-α expression, which triggers apoptosis of VCR-treated U937 and HL-60 cells. We also demonstrate that the apoptotic signalling is not affected by VCR-elicited microtubule destabilization.
Collapse
Affiliation(s)
- Liang-Jun Wang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chia-Hui Huang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yi-Jun Shi
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
29
|
Tian C, Chen X, Zhang Z, Wang X, Liu J. Design and synthesis of (2-(phenylamino)thieno[3,2-d]pyrimidin-4-yl)(3,4,5-trimethoxyphenyl)methanone analogues as potent anti-tubulin polymerization agents. Eur J Med Chem 2019; 183:111679. [DOI: 10.1016/j.ejmech.2019.111679] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/01/2022]
|
30
|
Delgado-Carreño C, Méndez-Callejas G. Topological properties and in vitro identification of essential nodes of the Paclitaxel and Vincristine interactomes in PC-3 cells. Biomed J 2019; 42:307-316. [PMID: 31783991 PMCID: PMC6888721 DOI: 10.1016/j.bj.2019.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/20/2022] Open
Abstract
Background Microtubule-targeting agents (MTAs) disrupt microtubule dynamics, thereby inducing apoptosis via mitochondrial pathway activation through the modulation in the expression of the Bcl-2 family. Methods To describe topological features of the MTAs networks associated to intrinsic apoptosis induction in p53-null prostate cancer cells, we predicted and compared the interactomes and topological properties of Paclitaxel and Vincristine, and thus, the essential nodes corresponding with the pro- and anti-apoptotic proteins and their kinetics were subjected to experimental analysis in PC-3 cell line. Results The essential nodes of the apoptotic pathways, TP53, and CASP3, were identified in both, Paclitaxel and Vincristine networks, but the intrinsic pathway markers BCL2, BAX, and BCL2L1 were identified as hub nodes only in the Paclitaxel network. An in vitro analysis demonstrated an increase in BimEL and the cleaved-caspase-3 proteins in PC-3 cells exposed to both treatments. Immunoprecipitation analysis showed that treatments induced the releasing of Bax from the anti-apoptotic complex with Bcl-2 protein and the role of BimEL as a de-repressor from sequestering complexes, in addition, new protein complexes were identified between BimEL or Bcl-2 and cleaved-caspase-3, contributing data to the Vincristine network for p53-null cells in response to MTAs. Conclusion The differences in sensitivities, protein profiles, and protein complex kinetics observed between the drugs confirmed that the selectivity and stimulation of the apoptotic system vary depending on the cell's genotype, the drug used and its exposure period.
Collapse
Affiliation(s)
- Claudia Delgado-Carreño
- Group of Biomedical Research and Applied Human Genetics, Laboratory of Cellular and Molecular Biology, School of Medicine, University of Applied and Environmental Sciences, U.D.C.A, Bogota, Colombia; Department of Chemistry, Faculty of Science, Javeriana University, Bogota, Colombia
| | - Gina Méndez-Callejas
- Group of Biomedical Research and Applied Human Genetics, Laboratory of Cellular and Molecular Biology, School of Medicine, University of Applied and Environmental Sciences, U.D.C.A, Bogota, Colombia.
| |
Collapse
|
31
|
Lin MS, Hong TM, Chou TH, Yang SC, Chung WC, Weng CW, Tsai ML, Cheng TJR, Chen JJW, Lee TC, Wong CH, Chein RJ, Yang PC. 4(1H)-quinolone derivatives overcome acquired resistance to anti-microtubule agents by targeting the colchicine site of β-tubulin. Eur J Med Chem 2019; 181:111584. [PMID: 31419740 DOI: 10.1016/j.ejmech.2019.111584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
Abstract
Developing new therapeutic strategies to overcome drug resistance of cancer cells is an ongoing endeavor. From among 2 million chemicals, we identified ethyl 4-oxo-2-phenyl-1,4-dihydroquinoline-6-carboxylate (AS1712) as a low-toxicity inhibitor of lung cancer cell proliferation and xenograft tumor growth. We show that AS1712 is active against broad cancer cell lines and is able to bind in the colchicine-binding pocket of β-tubulin, thereby inhibiting microtubule assembly and, consequently, inducing mitotic arrest and apoptosis. Our cell-based structure-activity relationship study identified a new lead compound, RJ-LC-15-8, which had a greater anti-proliferative potency for H1975 cells than did AS1712, while maintaining a similar mechanism of action. Notably, AS1712 and RJ-LC-15-8 overcame P-glycoprotein efflux pump and β-tubulin alterations that lead to acquired resistance against microtubule-targeting drugs of cancer cells. AS1712 and RJ-LC-15-8 may be lead compounds that overcome acquired resistance to microtubule-targeting agents of cancer cells.
Collapse
Affiliation(s)
- Ming-Shiu Lin
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Tse-Ming Hong
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ting-Hung Chou
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Shuenn-Chen Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Wei-Chia Chung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Chia-Wei Weng
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, 402, Taiwan
| | - Mei-Ling Tsai
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | | | - Jeremy J W Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, 402, Taiwan
| | - Te-Chang Lee
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Chi-Huey Wong
- The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Rong-Jie Chein
- Institute of Chemistry, Academia Sinica, Taipei, 115, Taiwan.
| | - Pan-Chyr Yang
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
32
|
Romagnoli R, Oliva P, Salvador MK, Camacho ME, Padroni C, Brancale A, Ferla S, Hamel E, Ronca R, Grillo E, Bortolozzi R, Rruga F, Mariotto E, Viola G. Design, synthesis and biological evaluation of novel vicinal diaryl-substituted 1H-Pyrazole analogues of combretastatin A-4 as highly potent tubulin polymerization inhibitors. Eur J Med Chem 2019; 181:111577. [PMID: 31400707 DOI: 10.1016/j.ejmech.2019.111577] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/17/2022]
Abstract
A series of 3-(3',4',5'-trimethoxyphenyl)-4-substituted 1H-pyrazole and their related 3-aryl-4-(3',4',5'-trimethoxyphenyl)-1-H-pyrazole regioisomeric derivatives, prepared as cis-rigidified combretastatin A-4 (CA-4) analogues, were synthesized and evaluated for their in vitro antiproliferative against six different cancer cell lines and, for selected highly active compounds, inhibitory effects on tubulin polymerization, cell cycle effects and in vivo potency. We retained the 3',4',5'-trimethoxyphenyl moiety as ring A throughout the present investigation, and a structure-activity relationship (SAR) information was obtained by adding electron-withdrawing (OCF3, CF3) or electron-releasing (alkyl and alkoxy) groups on the second aryl ring, corresponding to the B-ring of CA-4, either at the 3- or 4-position of the pyrazole nucleus. In addition, the B-ring was replaced with a benzo[b]thien-2-yl moiety. For many of the compounds, their activity was greater than, or comparable with, that of CA-4. Maximal activity was observed with the two regioisomeric derivatives characterized by the presence of a 4-ethoxyphenyl and a 3',4',5'-trimethoxyphenyl group at the C-3 and C-4 positions, and vice versa, of the 1H-pyrazole ring. The data showed that the 3',4',5'-trimethoxyphenyl moiety can be moved from the 3- to the 4-position of the 1H-pyrazole ring without significantly affecting antiproliferative activity. The most active derivatives bound to the colchicine site of tubulin and inhibited tubulin polymerization at submicromolar concentrations. In vivo experiments, on an orthotopic murine mammary tumor, revealed that 4c inhibited tumor growth even at low concentrations (5 mg/kg) compared to CA-4P (30 mg/kg).
Collapse
Affiliation(s)
- Romeo Romagnoli
- Dipartimento di Scienze Chimiche e Farmaceutiche, Via Luigi Borsari 46, Università di Ferrara, 44121, Ferrara, Italy.
| | - Paola Oliva
- Dipartimento di Scienze Chimiche e Farmaceutiche, Via Luigi Borsari 46, Università di Ferrara, 44121, Ferrara, Italy
| | - Maria Kimatrai Salvador
- Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain
| | - Maria Encarnacion Camacho
- Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain
| | - Chiara Padroni
- Aptuit, an Evotec Company, Via A. Fleming 4, 37135, Verona, Italy
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Salvatore Ferla
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Roberto Ronca
- Dipartimento di Medicina Molecolare e Traslazionale Unità di Oncologia Sperimentale ed Immunologia, Università di Brescia, 25123, Brescia, Italy
| | - Elisabetta Grillo
- Dipartimento di Medicina Molecolare e Traslazionale Unità di Oncologia Sperimentale ed Immunologia, Università di Brescia, 25123, Brescia, Italy
| | - Roberta Bortolozzi
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131, Padova, Italy
| | - Fatlum Rruga
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131, Padova, Italy
| | - Elena Mariotto
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131, Padova, Italy
| | - Giampietro Viola
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, Università di Padova, 35131, Padova, Italy; Istituto di Ricerca Pediatrica (IRP), Corso Stati Uniti 4, 35128, Padova, Italy.
| |
Collapse
|
33
|
Bortolozzi R, Carta D, Prà MD, Antoniazzi G, Mattiuzzo E, Sturlese M, Di Paolo V, Calderan L, Moro S, Hamel E, Quintieri L, Ronca R, Viola G, Ferlin MG. Evaluating the effects of fluorine on biological properties and metabolic stability of some antitubulin 3-substituted 7-phenyl-pyrroloquinolinones. Eur J Med Chem 2019; 178:297-314. [PMID: 31195171 DOI: 10.1016/j.ejmech.2019.05.092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 12/31/2022]
Abstract
A small number of fluorinated 7-phenyl-pyrroloquinolinone (7-PPyQ) derivatives was synthesized in an attempt to improve the metabolic stability of 3N-ethyl-7-PPyQ and 3N-benzoyl-7-PPyQ. The possible impacts of the fluorine-hydrogen isosterism on both biological activity and metabolic stability were evaluated. Introduction of a fluorine atom in the 2 or 3 position of the 7-phenyl ring yielded the 7-PPyQ derivatives 12, 13 and 15, which showed potent cytotoxicity (low micromolar and sub-nanomolar GI50s) both in human leukemic and solid tumor cell lines. None of them induced significant cell death in quiescent and proliferating human lymphocytes. Moreover, 12, 13 and 15 exhibited remarkable cytotoxic activity in the multidrug-resistant cell line CEMVbl100, suggesting that they are not substrates for P-glycoprotein. All compounds inhibited tubulin assembly and the binding of [3H]colchicine to tubulin, with the best activity occurring with compound 15. Mechanistic studies carried out on compound 12 indicated that it caused (a) a strong G2/M arrest; (b) apoptosis in a time- and concentration-dependent manner; (c) a significant production of ROS (in good agreement with the observed mitochondrial depolarization); (d) caspase-3 and poly (ADP-ribose) polymerase activation; and (e) a decrease in the expression of anti-apoptotic proteins. In vivo experiments in a murine syngeneic tumor model demonstrated that compounds 12 and 15 significantly reduced tumor mass at doses four times lower than that required for the reference compound combretastatin A-4 phosphate. Neither monofluorination of the 7-phenyl ring of 3N-ethyl-7-PPyQ nor replacement of the benzoyl function of 3N-benzoyl-7-PPyQ with a 2-fluorobenzoyl moiety led to any improvement in the metabolic stability.
Collapse
Affiliation(s)
- Roberta Bortolozzi
- Department of Woman's and Child's Health, University of Padova, Laboratory of Oncohematology, 35128, Padova, Italy
| | - Davide Carta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Matteo Dal Prà
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Giuseppe Antoniazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Elena Mattiuzzo
- Department of Woman's and Child's Health, University of Padova, Laboratory of Oncohematology, 35128, Padova, Italy
| | - Mattia Sturlese
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Veronica Di Paolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Laura Calderan
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Stefano Moro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Luigi Quintieri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, Oncology and Immunology Section, University of Brescia, 29881, Brescia, Italy
| | - Giampietro Viola
- Department of Woman's and Child's Health, University of Padova, Laboratory of Oncohematology, 35128, Padova, Italy
| | - Maria Grazia Ferlin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
34
|
Neagu M, Constantin C, Popescu ID, Zipeto D, Tzanakakis G, Nikitovic D, Fenga C, Stratakis CA, Spandidos DA, Tsatsakis AM. Inflammation and Metabolism in Cancer Cell-Mitochondria Key Player. Front Oncol 2019; 9:348. [PMID: 31139559 PMCID: PMC6527883 DOI: 10.3389/fonc.2019.00348] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/15/2019] [Indexed: 12/17/2022] Open
Abstract
Cancer metabolism is an essential aspect of tumorigenesis, as cancer cells have increased energy requirements in comparison to normal cells. Thus, an enhanced metabolism is needed in order to accommodate tumor cells' accelerated biological functions, including increased proliferation, vigorous migration during metastasis, and adaptation to different tissues from the primary invasion site. In this context, the assessment of tumor cell metabolic pathways generates crucial data pertaining to the mechanisms through which tumor cells survive and grow in a milieu of host defense mechanisms. Indeed, various studies have demonstrated that the metabolic signature of tumors is heterogeneous. Furthermore, these metabolic changes induce the exacerbated production of several molecules, which result in alterations that aid an inflammatory milieu. The therapeutic armentarium for oncology should thus include metabolic and inflammation regulators. Our expanding knowledge of the metabolic behavior of tumor cells, whether from solid tumors or hematologic malignancies, may provide the basis for the development of tailor-made cancer therapies.
Collapse
Affiliation(s)
- Monica Neagu
- Immunology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania.,Doctoral School, Biology Faculty, University of Bucharest, Bucharest, Romania.,Pathology Department, Colentina Clinical Hospital, Bucharest, Romania
| | - Carolina Constantin
- Immunology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania.,Pathology Department, Colentina Clinical Hospital, Bucharest, Romania
| | - Iulia Dana Popescu
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Donato Zipeto
- Department Neuroscience, Biomedicine and Movement Science, School of Medicine, University of Verona, Verona, Italy
| | - George Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Concettina Fenga
- Biomedical, Odontoiatric, Morphological and Functional Images Department, Occupational Medicine Section, University of Messina, Messina, Italy
| | - Constantine A Stratakis
- Section on Genetics & Endocrinology (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Greece
| | - Aristidis M Tsatsakis
- Department of Forensic Sciences and Toxicology, University of Crete, Heraklion, Greece
| |
Collapse
|
35
|
Synthesis, in vitro and in vivo biological evaluation of substituted 3-(5-imidazo[2,1-b]thiazolylmethylene)-2-indolinones as new potent anticancer agents. Eur J Med Chem 2019; 166:514-530. [PMID: 30784885 DOI: 10.1016/j.ejmech.2019.01.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/20/2018] [Accepted: 01/19/2019] [Indexed: 11/22/2022]
Abstract
A small library of 3-(5-imidazo[2,1-b]thiazolylmethylene)-2-indolinones has been synthesized and screened according to protocols available at the National Cancer Institute (NCI). Some derivatives were potent antiproliferative agents, showing GI50 values in the nanomolar range. Remarkably, when most active compounds against leukemia cells were tested in human peripheral blood lymphocytes from healthy donors, were 100-200 times less cytotoxic. Some compounds, selected by the Biological Evaluation Committee of NCI, were examined to determine tubulin assembly inhibition. Furthermore, flow cytometric studies performed on HeLa, HT-29, and A549 cells, showed that compounds 14 and 25 caused a block in the G2/M phase. Interestingly, these derivatives induced apoptosis through the mitochondrial death pathway, causing in parallel significant activation of both caspase-3 and -9, PARP cleavage and down-regulation of the anti-apoptotic proteins Bcl-2 and Mcl-1. Finally, compound 25 was also tested in vivo in the murine BL6-B16 melanoma and E0771 breast cancer cells, causing in both cases a significant reduction in tumor volume.
Collapse
|
36
|
Romagnoli R, Prencipe F, Oliva P, Baraldi S, Baraldi PG, Schiaffino Ortega S, Chayah M, Kimatrai Salvador M, Lopez-Cara LC, Brancale A, Ferla S, Hamel E, Ronca R, Bortolozzi R, Mariotto E, Mattiuzzo E, Viola G. Design, Synthesis, and Biological Evaluation of 6-Substituted Thieno[3,2- d]pyrimidine Analogues as Dual Epidermal Growth Factor Receptor Kinase and Microtubule Inhibitors. J Med Chem 2019; 62:1274-1290. [PMID: 30633509 DOI: 10.1021/acs.jmedchem.8b01391] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The clinical evidence for the success of tyrosine kinase inhibitors in combination with microtubule-targeting agents prompted us to design and develop single agents that possess both epidermal growth factor receptor (EGFR) kinase and tubulin polymerization inhibitory properties. A series of 6-aryl/heteroaryl-4-(3',4',5'-trimethoxyanilino)thieno[3,2- d]pyrimidine derivatives were discovered as novel dual tubulin polymerization and EGFR kinase inhibitors. The 4-(3',4',5'-trimethoxyanilino)-6-( p-tolyl)thieno[3,2- d]pyrimidine derivative 6g was the most potent compound of the series as an antiproliferative agent, with half-maximal inhibitory concentration (IC50) values in the single- or double-digit nanomolar range. Compound 6g bound to tubulin in the colchicine site and inhibited tubulin assembly with an IC50 value of 0.71 μM, and 6g inhibited EGFR activity with an IC50 value of 30 nM. Our data suggested that the excellent in vitro and in vivo profile of 6g may be derived from its dual inhibition of tubulin polymerization and EGFR kinase.
Collapse
Affiliation(s)
- Romeo Romagnoli
- Dipartimento di Scienze Chimiche e Farmaceutiche , Università degli Studi di Ferrara , Via Luigi Borsari 46 , 44121 Ferrara , Italy
| | - Filippo Prencipe
- Dipartimento di Scienze Chimiche e Farmaceutiche , Università degli Studi di Ferrara , Via Luigi Borsari 46 , 44121 Ferrara , Italy
| | - Paola Oliva
- Dipartimento di Scienze Chimiche e Farmaceutiche , Università degli Studi di Ferrara , Via Luigi Borsari 46 , 44121 Ferrara , Italy
| | - Stefania Baraldi
- Dipartimento di Scienze Chimiche e Farmaceutiche , Università degli Studi di Ferrara , Via Luigi Borsari 46 , 44121 Ferrara , Italy
| | - Pier Giovanni Baraldi
- Dipartimento di Scienze Chimiche e Farmaceutiche , Università degli Studi di Ferrara , Via Luigi Borsari 46 , 44121 Ferrara , Italy
| | - Santiago Schiaffino Ortega
- Departamento de Química Farmacéutica y Orgánica , Facultad de Farmacia , Campus de Cartuja s/n , 18071 Granada , Spain
| | - Mariem Chayah
- Departamento de Química Farmacéutica y Orgánica , Facultad de Farmacia , Campus de Cartuja s/n , 18071 Granada , Spain
| | - Maria Kimatrai Salvador
- Departamento de Química Farmacéutica y Orgánica , Facultad de Farmacia , Campus de Cartuja s/n , 18071 Granada , Spain
| | - Luisa Carlota Lopez-Cara
- Departamento de Química Farmacéutica y Orgánica , Facultad de Farmacia , Campus de Cartuja s/n , 18071 Granada , Spain
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences , Cardiff University , King Edward VII Avenue , Cardiff CF10 3NB , U.K
| | - Salvatore Ferla
- School of Pharmacy and Pharmaceutical Sciences , Cardiff University , King Edward VII Avenue , Cardiff CF10 3NB , U.K
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research , National Cancer Institute, National Institutes of Health , Frederick , Maryland 21702 , United States
| | - Roberto Ronca
- Dipartimento di Medicina Molecolare e Traslazionale Unità di Oncologia Sperimentale ed Immunologia , Università di Brescia , 25123 Brescia , Italy
| | - Roberta Bortolozzi
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia , Università di Padova , 35131 Padova , Italy
| | - Elena Mariotto
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia , Università di Padova , 35131 Padova , Italy
| | - Elena Mattiuzzo
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia , Università di Padova , 35131 Padova , Italy
| | - Giampietro Viola
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia , Università di Padova , 35131 Padova , Italy.,Istituto di Ricerca Pediatrica (IRP) , Corso Stati Uniti 4 , 35128 Padova , Italy
| |
Collapse
|
37
|
Sato-Kaneko F, Wang X, Yao S, Hosoya T, Lao FS, Messer K, Pu M, Shukla NM, Cottam HB, Chan M, Carson DA, Corr M, Hayashi T. Discovery of a Novel Microtubule Targeting Agent as an Adjuvant for Cancer Immunotherapy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8091283. [PMID: 30406141 PMCID: PMC6199861 DOI: 10.1155/2018/8091283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/17/2018] [Indexed: 12/26/2022]
Abstract
For an activating immunotherapy such as adjuvants, a compound that can prolong immune stimulation may enhance efficacy. We leveraged data from two prior high throughput screens with NF-κB and interferon reporter cell lines to identify 4H-chromene-3-carbonitriles as a class of compounds that prolonged activation in both screens. We repurchased 23 of the most promising candidates. Out of these compounds we found #1 to be the most effective agent in stimulating the release of cytokines and chemokines from immune cells, including murine primary bone marrow derived dendritic cells. Mechanistically, #1 inhibited tubulin polymerization, and its effect on immune cell activation was abolished in cells mutated in the beta-tubulin gene (TUBB) encoding the site where colchicine binds. Treatment with #1 resulted in mitochondrial depolarization followed by mitogen-activated protein kinase activation. Because tubulin polymerization modulating agents have been used for chemotherapy to treat malignancy and #1 activated cytokine responses, we hypothesized that #1 could be effective for cancer immunotherapy. Intratumoral injection of #1 delayed tumor growth in a murine syngeneic model of head and neck cancer. When combined with PD-1 blockade, tumor growth slowed in the injected tumor nodule and there was an abscopal effect in an uninjected nodule on the contralateral flank, suggesting central antitumor immune activation. Thus, we identified a new class of tubulin depolymerizing agent that acts as both an innate and an adaptive immune activating agent and that limits solid tumor growth when used concurrently with a checkpoint inhibitor.
Collapse
Affiliation(s)
- Fumi Sato-Kaneko
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Xiaodong Wang
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Nanhai Ave 3688, Shenzhen, Guangdong 518060, China
| | - Shiyin Yao
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Tadashi Hosoya
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Fitzgerald S. Lao
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Karen Messer
- Division of Biostatistics, University of California San Diego, La Jolla 92093, USA
| | - Minya Pu
- Division of Biostatistics, University of California San Diego, La Jolla 92093, USA
| | - Nikunj M. Shukla
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Howard B. Cottam
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Michael Chan
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Dennis A. Carson
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla 92093, USA
| | - Tomoko Hayashi
- Moores Cancer Center, University of California San Diego, La Jolla 92093, USA
| |
Collapse
|
38
|
Takac P, Kello M, Pilatova MB, Kudlickova Z, Vilkova M, Slepcikova P, Petik P, Mojzis J. New chalcone derivative exhibits antiproliferative potential by inducing G2/M cell cycle arrest, mitochondrial-mediated apoptosis and modulation of MAPK signalling pathway. Chem Biol Interact 2018; 292:37-49. [PMID: 29981726 DOI: 10.1016/j.cbi.2018.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/04/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023]
Abstract
In the present study, we investigated antiproliferative activity of seven newly synthesized chalcone derivatives. Among tested compounds, (2 E)-3-(acridin-9-yl)-1-(2,6-dimethoxyphenyl)prop-2-en-1-one (1C) was the most potent with IC50 = 4.1 μmol/L in human colorectal HCT116 cells and was selected for further studies. Inhibition of cell proliferation was associated with cell cycle arrest in G2/M phase and dysregulation of α, α1 and β5 tubulins. Moreover, 1C caused disruption of the mitochondrial membrane potential and increased number of cells with sub G0/G1 DNA content which is considered as marker of apoptosis. Apoptosis was confirmed by annexin V/PI and AO/PI staining. Furthermore, we found increased concentration of cytochrome c, Smac/Diablo and increased caspase-3 and caspase-9 activity, cleavage of PARP as well as activation of DNA repair mechanisms in 1C-treated HCT116 cancer cells. Moreover this chalcone derivative up-regulated proapoptotic Bax expression and down-regulated antiapoptotic Bcl-2 and Bcl-xL expression. Additionally, 1C treatment led to modulation of MAPKs and Akt signalling pathways. In conclusion, our data showed ability of 1C to suppress cancel cell growth and provide the rationale for further in vivo study.
Collapse
Affiliation(s)
- Peter Takac
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, 04011, Kosice, Slovak Republic
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, 04011, Kosice, Slovak Republic
| | - Martina Bago Pilatova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, 04011, Kosice, Slovak Republic
| | - Zuzana Kudlickova
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Safarik University, 040 01, Kosice, Slovak Republic; Department of Chemistry, Biochemistry and Biophysics, University of Veterinary Medicine and Pharmacy, 04181, Košice, Slovak Republic
| | - Maria Vilkova
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Safarik University, 040 01, Kosice, Slovak Republic
| | - Pavlina Slepcikova
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Safarik University, 040 01, Kosice, Slovak Republic
| | - Peter Petik
- Department of Pathology, Pavol Jozef Safarik University, 040 01, Kosice, Slovak Republic
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, 04011, Kosice, Slovak Republic.
| |
Collapse
|
39
|
Molecular basis of resistance to the microtubule-depolymerizing antitumor compound plocabulin. Sci Rep 2018; 8:8616. [PMID: 29872155 PMCID: PMC5988728 DOI: 10.1038/s41598-018-26736-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/17/2018] [Indexed: 12/13/2022] Open
Abstract
Plocabulin (PM060184) is a microtubule depolymerizing agent with potent antiproliferative activity undergoing phase II clinical trials for the treatment of solid tumors. Plocabulin shows antifungal activity virtually abolishing growth of the filamentous fungus Aspergillus nidulans. A. nidulans hyphae depend both on mitotic and interphase microtubules, as human cells. Here, we exploited the A. nidulans genetic amenability to gain insight into the mechanism of action of plocabulin. By combining mutations in the two A. nidulans β-tubulin isotypes we obtained a plocabulin-insensitive strain, showing that β-tubulin is the only molecular target of plocabulin in fungal cells. From a genetic screen, we recovered five mutants that show plocabulin resistance but do not carry mutations in β-tubulin. Resistance mutations resulted in amino acid substitutions in (1) two subunits of the eukaryotic translation initiation factor eIF2B activating the General Amino Acid Control, (2) TIM44, an essential component of the inner mitochondrial membrane translocase, (3) two transcription factors of the binuclear zinc cluster family potentially interfering with the uptake or efflux of plocabulin. Given the conservation of some of the identified proteins and their respective cellular functions in the tumor environment, our results pinpoint candidates to be tested as potential biomarkers for determination of drug efficiency.
Collapse
|
40
|
Abstract
Background Despite chemotherapy intensification, a subgroup of high-risk paediatric T-cell acute lymphoblastic leukemia (T-ALL) patients still experience treatment failure. In this context, we hypothesised that therapy resistance in T-ALL might involve aldo-keto reductase 1C (AKR1C) enzymes as previously reported for solid tumors. Methods Expression of NRF2-AKR1C signaling components has been analysed in paediatric T-ALL samples endowed with different treatment outcomes as well as in patient-derived xenografts of T-ALL. The effects of AKR1C enzyme modulation has been investigated in T-ALL cell lines and primary cultures by combining AKR1C inhibition, overexpression, and gene silencing approaches. Results We show that T-ALL cells overexpress AKR1C1-3 enzymes in therapy-resistant patients. We report that AKR1C1-3 enzymes play a role in the response to vincristine (VCR) treatment, also ex vivo in patient-derived xenografts. Moreover, we demonstrate that the modulation of AKR1C1-3 levels is sufficient to sensitise T-ALL cells to VCR. Finally, we show that T-ALL chemotherapeutics induce overactivation of AKR1C enzymes independent of therapy resistance, thus establishing a potential resistance loop during T-ALL combination treatment. Conclusions Here, we demonstrate that expression and activity of AKR1C enzymes correlate with response to chemotherapeutics in T-ALL, posing AKR1C1-3 as potential targets for combination treatments during T-ALL therapy.
Collapse
|
41
|
Romagnoli R, Kimatrai Salvador M, Schiaffino Ortega S, Baraldi PG, Oliva P, Baraldi S, Lopez-Cara LC, Brancale A, Ferla S, Hamel E, Balzarini J, Liekens S, Mattiuzzo E, Basso G, Viola G. 2-Alkoxycarbonyl-3-arylamino-5-substituted thiophenes as a novel class of antimicrotubule agents: Design, synthesis, cell growth and tubulin polymerization inhibition. Eur J Med Chem 2018; 143:683-698. [PMID: 29220790 DOI: 10.1016/j.ejmech.2017.11.096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/12/2017] [Accepted: 11/29/2017] [Indexed: 11/30/2022]
Abstract
Microtubules are recognized as crucial components of the mitotic spindle during cell division, and, for this reason, the microtubule system is an attractive target for the development of anticancer agents. Continuing our search strategy for novel tubulin targeting-compounds, a new series of 2-alkoxycarbonyl-3-(3',4',5'-trimethoxyanilino)-5-aryl/heteroarylthiophene derivatives was designed, synthesized and demonstrated to act as tubulin polymerization inhibitors at the colchicine site. A structure-activity relationship study on the phenyl at the 5-position of the thiophene ring was performed by introducing a variety of substituents containing electron-releasing and electron-withdrawing groups, with the 2-alkoxycarbonyl-3-(3',4',5'-trimethoxyanilino)thiophene scaffold being the minimum structural requirement for activity. Of the tested compounds, derivatives 4a, 4c, 4i and 4k possessed the highest overall potency and displayed high antiproliferative activities at submicromolar concentrations, with IC50 values ranging from 0.13 to 0.84 μM against four different cancer cell lines. Three agents (4a, 4c and 4i) in the present series had similar effects, and these were comparable to those of the reference compound combretastatin A-4 (CA-4) as inhibitors of tubulin assembly. The antitubulin effects correlated with the cytostatic activities and indicate that these compounds inhibit cell growth through inhibition of tubulin polymerization by binding at the colchicine site. Compound 4c, containing the 2'-thienyl ring at the 5-position of the 2-methoxycarbonyl-3-(3',4',5'-trimethoxyanilino)thiophene scaffold, exhibited substantial antiproliferative activity with a mean IC50 value of 140 nM, inhibited tubulin polymerization with an IC50 value of 1.2 μM, similar to that of CA-4 (IC50: 1.1 μM), and induced apoptosis in HeLa cells.
Collapse
Affiliation(s)
- Romeo Romagnoli
- Dipartimento di Scienze Chimiche e Farmaceutiche, Università di Ferrara, 44121 Ferrara, Italy.
| | - Maria Kimatrai Salvador
- Departamento de Química Farmaceútica y Orgánica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain
| | - Santiago Schiaffino Ortega
- Departamento de Química Farmaceútica y Orgánica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain
| | - Pier Giovanni Baraldi
- Dipartimento di Scienze Chimiche e Farmaceutiche, Università di Ferrara, 44121 Ferrara, Italy
| | - Paola Oliva
- Dipartimento di Scienze Chimiche e Farmaceutiche, Università di Ferrara, 44121 Ferrara, Italy
| | - Stefania Baraldi
- Dipartimento di Scienze Chimiche e Farmaceutiche, Università di Ferrara, 44121 Ferrara, Italy
| | - Luisa Carlota Lopez-Cara
- Departamento de Química Farmaceútica y Orgánica, Facultad de Farmacia, Campus de Cartuja s/n, 18071, Granada, Spain
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Salvatore Ferla
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Jan Balzarini
- Rega Institute for Medical Research, KU Leuven, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | - Sandra Liekens
- Rega Institute for Medical Research, KU Leuven, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | - Elena Mattiuzzo
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia Pediatrica, Università di Padova, 35131 Padova, Italy
| | - Giuseppe Basso
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia Pediatrica, Università di Padova, 35131 Padova, Italy
| | - Giampietro Viola
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia Pediatrica, Università di Padova, 35131 Padova, Italy.
| |
Collapse
|
42
|
Targeting tubulin polymerization by novel 7-aryl-pyrroloquinolinones: Synthesis, biological activity and SARs. Eur J Med Chem 2017; 143:244-258. [PMID: 29197729 DOI: 10.1016/j.ejmech.2017.11.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 01/21/2023]
Abstract
Earlier studies had confirmed that the 7-phenylpyrroloquinolinone (7-PPyQ) nucleus was an important scaffold for new chemotherapeutic drugs targeting microtubules. For wide-ranging SARs, a series of derivatives were synthesized through a robust procedure. For comparison with the reference 3-ethyl-7-PPyQ 31, the angular geometry and substituents at the 3 and 7 positions were varied to explore interactions inside the colchicine site of tubulin. Of the new compounds synthesized, potent cytotoxicity (low and sub-nanomolar GI50 values) was observed with 21 and 24, both more potent than 31, in both leukemic and solid tumor cell lines. Neither compound 21 nor 24 induced significant cell death in normal human lymphocytes, suggesting that the compounds may be selectively active against cancer cells. In particular, 24 was a potent inducer of apoptosis in the A549 and HeLa cell lines. With both compounds, induction of apoptosis was associated with dissipation of the mitochondrial transmembrane potential and production of reactive oxygen species, indicating that cells treated with the compounds followed the intrinsic pathway of apoptosis. Moreover, immunoblot analysis revealed that compound 24 even at 50 nM reduced the expression of anti-apoptotic proteins such as Bcl-2 and Mcl-1. Finally, molecular docking studies of the newly synthesized compounds demonstrate that active pyrroloquinolinone derivatives strongly bind in the colchicine site of β-tubulin.
Collapse
|
43
|
Parker AL, Teo WS, McCarroll JA, Kavallaris M. An Emerging Role for Tubulin Isotypes in Modulating Cancer Biology and Chemotherapy Resistance. Int J Mol Sci 2017; 18:ijms18071434. [PMID: 28677634 PMCID: PMC5535925 DOI: 10.3390/ijms18071434] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 06/24/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022] Open
Abstract
Tubulin proteins, as components of the microtubule cytoskeleton perform critical cellular functions throughout all phases of the cell cycle. Altered tubulin isotype composition of microtubules is emerging as a feature of aggressive and treatment refractory cancers. Emerging evidence highlighting a role for tubulin isotypes in differentially influencing microtubule behaviour and broader functional networks within cells is illuminating a complex role for tubulin isotypes regulating cancer biology and chemotherapy resistance. This review focuses on the role of different tubulin isotypes in microtubule dynamics as well as in oncogenic changes that provide a survival or proliferative advantage to cancer cells within the tumour microenvironment and during metastatic processes. Consideration of the role of tubulin isotypes beyond their structural function will be essential to improving the current clinical use of tubulin-targeted chemotherapy agents and informing the development of more effective cancer therapies.
Collapse
Affiliation(s)
- Amelia L Parker
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Wee Siang Teo
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Joshua A McCarroll
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Maria Kavallaris
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
44
|
Hoogerheide DP, Noskov SY, Jacobs D, Bergdoll L, Silin V, Worcester DL, Abramson J, Nanda H, Rostovtseva TK, Bezrukov SM. Structural features and lipid binding domain of tubulin on biomimetic mitochondrial membranes. Proc Natl Acad Sci U S A 2017; 114:E3622-E3631. [PMID: 28420794 PMCID: PMC5422764 DOI: 10.1073/pnas.1619806114] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dimeric tubulin, an abundant water-soluble cytosolic protein known primarily for its role in the cytoskeleton, is routinely found to be associated with mitochondrial outer membranes, although the structure and physiological role of mitochondria-bound tubulin are still unknown. There is also no consensus on whether tubulin is a peripheral membrane protein or is integrated into the outer mitochondrial membrane. Here the results of five independent techniques-surface plasmon resonance, electrochemical impedance spectroscopy, bilayer overtone analysis, neutron reflectometry, and molecular dynamics simulations-suggest that α-tubulin's amphipathic helix H10 is responsible for peripheral binding of dimeric tubulin to biomimetic "mitochondrial" membranes in a manner that differentiates between the two primary lipid headgroups found in mitochondrial membranes, phosphatidylethanolamine and phosphatidylcholine. The identification of the tubulin dimer orientation and membrane-binding domain represents an essential step toward our understanding of the complex mechanisms by which tubulin interacts with integral proteins of the mitochondrial outer membrane and is important for the structure-inspired design of tubulin-targeting agents.
Collapse
Affiliation(s)
- David P Hoogerheide
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD 20899;
| | - Sergei Y Noskov
- Center for Molecular Simulations, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada T2N 1N4;
| | - Daniel Jacobs
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Lucie Bergdoll
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Vitalii Silin
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD 20899
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - David L Worcester
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD 20899
| | - Jeff Abramson
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, Karnataka, India
| | - Hirsh Nanda
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD 20899
- Physics Department, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Tatiana K Rostovtseva
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Sergey M Bezrukov
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
45
|
Lai SR, Castello SA, Robinson AC, Koehler JW. In vitro anti-tubulin effects of mebendazole and fenbendazole on canine glioma cells. Vet Comp Oncol 2017; 15:1445-1454. [PMID: 28078780 DOI: 10.1111/vco.12288] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 10/12/2016] [Accepted: 10/13/2016] [Indexed: 01/19/2023]
Abstract
Benzimidazole anthelmintics have reported anti-neoplastic effects both in vitro and in vivo. The purpose of this study was to evaluate the in vitro chemosensitivity of three canine glioma cell lines to mebendazole and fenbendazole. The mean inhibitory concentration (IC50 ) (±SD) obtained from performing the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay after treating J3T, G06-A, and SDT-3G cells for 72 h with mebendazole were 0.030 ± 0.003, 0.080 ± 0.015 and 0.030 ± 0.006 μM respectively, while those for fenbendazole were 0.550 ± 0.015, 1.530 ± 0.159 and 0.690 ± 0.095 μM; treatment of primary canine fibroblasts for 72 h at IC50 showed no significant effect. Immunofluorescence studies showed disruption of tubulin after treatment. Mebendazole and fenbendazole are cytotoxic in canine glioma cell lines in vitro and may be good candidates for treatment of canine gliomas. Further in vivo studies are required.
Collapse
Affiliation(s)
- S R Lai
- Department of Clinical Sciences, Auburn University College of Veterinary Medicine, Auburn, AL, USA
| | - S A Castello
- Undergraduate Honors College, Auburn University, Auburn, AL, USA
| | - A C Robinson
- Undergraduate Honors College, Auburn University, Auburn, AL, USA
| | - J W Koehler
- Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, USA
| |
Collapse
|
46
|
Carta D, Bortolozzi R, Sturlese M, Salmaso V, Hamel E, Basso G, Calderan L, Quintieri L, Moro S, Viola G, Ferlin MG. Synthesis, structure-activity relationships and biological evaluation of 7-phenyl-pyrroloquinolinone 3-amide derivatives as potent antimitotic agents. Eur J Med Chem 2016; 127:643-660. [PMID: 27823884 DOI: 10.1016/j.ejmech.2016.10.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 12/20/2022]
Abstract
A small library of 7-pyrrolo[3,2-f]quinolinones was obtained by introducing benzoyl, sulfonyl and carbamoyl side chains at the 3-N position, and their cytotoxicity against a panel of leukemic and solid tumor cell lines was evaluated. Most of them showed high antiproliferative activity with GI50s ranging from micro-to sub-nanomolar values, and these values correlated well with the inhibitory activities of the compounds against tubulin polymerization. Based on a recently proposed colchicine bind site inhibitors (CBSIs) pharmacophore, the interactions of the novel 7-PPyQs at the colchicine domain were rationalized. The most active compounds (4a and 4b) did not induce significant cell death in normal human lymphocytes, suggesting that the compounds may be selective against cancer cells. In particular, 4a was a potent inducer of apoptosis in both the HeLa and Jurkat cell lines. On the other hand, the sulfonyl derivative 4b exhibited a lower potency in comparison with 4a. With both compounds, induction of apoptosis was associated with dissipation of the mitochondrial transmembrane potential and production of reactive oxygen species, suggesting that cells treated with the compounds followed the intrinsic pathway of apoptosis.
Collapse
Affiliation(s)
- Davide Carta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Roberta Bortolozzi
- Department of Woman's and Child's Health, University of Padova, Laboratory of Oncohematology, 35128, Padova, Italy
| | - Mattia Sturlese
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Veronica Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Giuseppe Basso
- Department of Woman's and Child's Health, University of Padova, Laboratory of Oncohematology, 35128, Padova, Italy
| | - Laura Calderan
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Luigi Quintieri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Stefano Moro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Giampietro Viola
- Department of Woman's and Child's Health, University of Padova, Laboratory of Oncohematology, 35128, Padova, Italy
| | - Maria Grazia Ferlin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
47
|
Philchenkov AA, Zavelevich MP, Tryndyak VP, Kuiava LM, Blokhin DY, Miura K, Silvestri R, Pogribny IP. Antiproliferative and proapoptotic effects of a pyrrole containing arylthioindole in human Jurkat leukemia cell line and multidrug-resistant Jurkat/A4 cells. Cancer Biol Ther 2016; 16:1820-9. [PMID: 26785947 DOI: 10.1080/15384047.2015.1078026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recently, a series of novel arylthioindole compounds, potent inhibitors of tubulin polymerization and cancer cell growth, were synthesized. In the present study the effects of 2-(1H-pyrrol-3-yl)-3-((3,4,5-trimethoxyphenyl)thio)-1H-indole (ATI5 compound) on cell proliferation, cell cycle progression, and induction of apoptosis in human T-cell acute leukemia Jurkat cells and their multidrug resistant Jurkat/A4 subline were investigated. Treatment of the Jurkat cells with the ATI5 compound for 48 hrs resulted in a strong G2/M cell cycle arrest and p53-independent apoptotic cell death accompanied by the induction of the active form of caspase-3 and poly(ADP-ribose) polymerase-1 (PARP-1) cleavage. ATI5 treatment also caused non-cell death related mitotic arrest in multidrug resistant Jurkat/A4 cells after 48 hrs of treatment suggesting promising opportunities for the further design of pyrrole-containing ATI compounds as anticancer agents. Cell death resistance of Jurkat/A4 cells to ATI5 compound was associated with alterations in the expression of pro-survival and anti-apoptotic protein-coding and microRNA genes. More importantly, findings showing that ATI5 treatment induced p53-independent apoptosis are of great importance from a therapeutic point of view since p53 mutations are common genetic alterations in human neoplasms.
Collapse
Affiliation(s)
- Alex A Philchenkov
- a R. E. Kavetsky Institute of Experimental Oncology, Pathology and Radiobiology; National Academy of Sciences of Ukraine ; Kyiv , Ukraine
| | - Michael P Zavelevich
- a R. E. Kavetsky Institute of Experimental Oncology, Pathology and Radiobiology; National Academy of Sciences of Ukraine ; Kyiv , Ukraine
| | - Volodymyr P Tryndyak
- b Division of Biochemical Toxicology; National Center for Toxicological Research ; Jefferson , AR USA
| | - Ludmila M Kuiava
- a R. E. Kavetsky Institute of Experimental Oncology, Pathology and Radiobiology; National Academy of Sciences of Ukraine ; Kyiv , Ukraine
| | - Dmitry Yu Blokhin
- c N. N. Blokhin Cancer Research Center ; Moscow , Russian Federation
| | - Koh Miura
- d Miyagi Cancer Center ; Natori , Japan
| | - Romano Silvestri
- e Istituto Pasteur-Fondazione Cenci Bolognetti; Sapienza Università di Roma ; Roma , Italy
| | - Igor P Pogribny
- b Division of Biochemical Toxicology; National Center for Toxicological Research ; Jefferson , AR USA
| |
Collapse
|
48
|
Bates D, Eastman A. Microtubule destabilising agents: far more than just antimitotic anticancer drugs. Br J Clin Pharmacol 2016; 83:255-268. [PMID: 27620987 DOI: 10.1111/bcp.13126] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/11/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
Vinca alkaloids have been approved as anticancer drugs for more than 50 years. They have been classified as cytotoxic chemotherapy drugs that act during cellular mitosis, enabling them to target fast growing cancer cells. With the evolution of cancer drug development there has been a shift towards new "targeted" therapies to avoid the side effects and general toxicities of "cytotoxic chemotherapies" such as the vinca alkaloids. Due to their original classification, many have overlooked the fact that vinca alkaloids, taxanes and related drugs do have a specific molecular target: tubulin. They continue to be some of the most effective anticancer drugs, perhaps because their actions upon the microtubule network extend far beyond the ability to halt cells in mitosis, and include the induction of apoptosis at all phases of the cell cycle. In this review, we highlight the numerous cellular consequences of disrupting microtubule dynamics, expanding the textbook knowledge of microtubule destabilising agents and providing novel opportunities for their use in cancer therapy.
Collapse
Affiliation(s)
- Darcy Bates
- Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Alan Eastman
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
49
|
Wei RJ, Lin SS, Wu WR, Chen LR, Li CF, Chen HD, Chou CT, Chen YC, Liang SS, Chien ST, Shiue YL. A microtubule inhibitor, ABT-751, induces autophagy and delays apoptosis in Huh-7 cells. Toxicol Appl Pharmacol 2016; 311:88-98. [PMID: 27678524 DOI: 10.1016/j.taap.2016.09.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 09/15/2016] [Accepted: 09/22/2016] [Indexed: 12/28/2022]
Abstract
The objective was to investigate the upstream mechanisms of apoptosis which were triggered by a novel anti-microtubule drug, ABT-751, in hepatocellular carcinoma-derived Huh-7 cells. Effects of ABT-751 were evaluated by immunocytochemistry, flow cytometric, alkaline comet, soft agar, immunoblotting, CytoID, green fluorescent protein-microtubule associated protein 1 light chain 3 beta detection, plasmid transfection, nuclear/cytosol fractionation, coimmunoprecipitation, quantitative reverse transcription-polymerase chain reaction, small-hairpin RNA interference and mitochondria/cytosol fractionation assays. Results showed that ABT-751 caused dysregulation of microtubule, collapse of mitochondrial membrane potential, generation of reactive oxygen species (ROS), DNA damage, G2/M cell cycle arrest, inhibition of anchorage-independent cell growth and apoptosis in Huh-7 cells. ABT-751 also induced early autophagy via upregulation of nuclear TP53 and downregulation of the AKT serine/threonine kinase (AKT)/mechanistic target of rapamycin (MTOR) pathway. Through modulation of the expression levels of DNA damage checkpoint proteins and G2/M cell cycle regulators, ABT-751 induced G2/M cell cycle arrest. Subsequently, ABT-751 triggered apoptosis with marked downregulation of B-cell CLL/lymphoma 2, upregulation of mitochondrial BCL2 antagonist/killer 1 and BCL2 like 11 protein levels, and cleavages of caspase 8 (CASP8), CASP9, CASP3 and DNA fragmentation factor subunit alpha proteins. Suppression of ROS significantly decreased ABT-751-induced autophagic and apoptotic cells. Pharmacological inhibition of autophagy significantly increased the percentages of ABT-751-induced apoptotic cells. The autophagy induced by ABT-751 plays a protective role to postpone apoptosis by exerting adaptive responses following microtubule damage, ROS and/or impaired mitochondria.
Collapse
Affiliation(s)
- Ren-Jie Wei
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Pathology, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Su-Shuan Lin
- Department of Pathology, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Wen-Ren Wu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Lih-Ren Chen
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan; Division of Physiology, Livestock Research Institute, Council of Agriculture, Taiwan
| | - Chien-Feng Li
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan; Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institute, Tainan, Taiwan; Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Han-De Chen
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chien-Ting Chou
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ya-Chun Chen
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Shih-Shin Liang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shang-Tao Chien
- Department of Pathology, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
50
|
Kuo TC, Li LW, Pan SH, Fang JM, Liu JH, Cheng TJ, Wang CJ, Hung PF, Chen HY, Hong TM, Hsu YL, Wong CH, Yang PC. Purine-Type Compounds Induce Microtubule Fragmentation and Lung Cancer Cell Death through Interaction with Katanin. J Med Chem 2016; 59:8521-34. [DOI: 10.1021/acs.jmedchem.6b00797] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Ting-Chun Kuo
- Ph.D.
Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei, Taiwan
| | - Ling-Wei Li
- Department
of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Szu-Hua Pan
- Ph.D.
Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei, Taiwan
- Graduate
Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Genome
and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 106, Taiwan
| | - Jim-Min Fang
- Department
of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Jyung-Hurng Liu
- Department
of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
- Institute
of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
- Agricultural
Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- Rong
Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Ting-Jen Cheng
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Jen Wang
- Department
of Internal Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Institute
of Stem Cell and Translational Cancer Research, Chang Gung Memorial HospitalTaipei 105, Taiwan
| | - Pei-Fang Hung
- Department
of Internal Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsuan-Yu Chen
- Institute
of Statistical Science, Academia Sinica, Taipei 115, Taiwan
| | - Tse-Ming Hong
- Institute
of Clinical Medicine, National Cheng Kung University College of Medicine, Tainan 701, Taiwan
| | - Yuan-Ling Hsu
- Graduate
Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chi-Huey Wong
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Pan-Chyr Yang
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- NTU
Center for Genomic Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|