1
|
Li J, Zhang T, Wu D, He C, Weng H, Zheng T, Liu J, Yao H, Chen J, Ren Y, Zhu Z, Xu J, Xu S. Palladium-Mediated Bioorthogonal System for Prodrug Activation of N-Benzylbenzamide-Containing Tubulin Polymerization Inhibitors for the Treatment of Solid Tumors. J Med Chem 2024. [PMID: 39484713 DOI: 10.1021/acs.jmedchem.4c02419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Bioorthogonal cleavage reactions have been developed as an intriguing strategy to enhance the safety of chemotherapeutics. Aiming to reduce the toxicity and improve the targeted release properties of the colchicine binding site inhibitors (CBSIs) based on previous work, a series of biologically inert prodrugs were further designed and synthesized through a bioorthogonal prodrug strategy. The therapeutic effects of prodrugs could be "turned-on" once combined with palladium resins. Particularly, prodrug 2b was 68.3-fold less cytotoxic compared to the parent compound, while its cytotoxicity was recovered in situ in the presence of palladium resins. Mechanism studies confirmed that 2b inhibited cell growth in the same manner as CBSIs. More importantly, in vivo efficacy studies demonstrated the efficient activation of 2b by palladium resins, resulting in significant inhibition of tumor growth (63.2%). These results suggest that prodrug 2b with improved safety and targeted release property catalyzed by a Pd-mediated bioorthogonal cleavage reaction deserves further investigation.
Collapse
Affiliation(s)
- Jinlong Li
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Tong Zhang
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Di Wu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Chen He
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Haoxiang Weng
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Tiandong Zheng
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Jie Liu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Hong Yao
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Jichao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yansong Ren
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, U.K
| | - Jinyi Xu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
| | - Shengtao Xu
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P.R. China
- Department of Hepatobiliary Surgery, The First People's Hospital of Kunshan, Suzhou 215132, P.R. China
| |
Collapse
|
2
|
Jin YZ, Xin YB, Li Y, Chen XY, Man DA, Tian YS. Synthesis and Selective Anticancer Activity Evaluation of 2-phenylacrylonitrile Derivatives as Tubulin Inhibitors. Curr Med Chem 2024; 31:2090-2106. [PMID: 38384112 PMCID: PMC11071649 DOI: 10.2174/0109298673263854231009063053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/17/2023] [Accepted: 09/01/2023] [Indexed: 02/23/2024]
Abstract
OBJECTIVE This study aimed at synthesizing 13 series of novel derivatives with 2-phenylacrylonitrile, evaluating antitumor activity both in vivo and in vitro, and obtaining novel tubulin inhibitors. METHOD The 13 series of 2-phenylacrylonitrile derivatives were synthesized by Knoevenagel condensation and the anti-proliferative activities were determined by MTT assay. The cell cycle and apoptosis were analyzed by flow cytometer. Quantitative cell migration was performed using 24-well Boyden chambers. The proteins were detected by western blotting. in vitro kinetics of microtubule assembly was measured using ELISA kit for Human β-tubulin (TUBB). Molecular docking was done by Discovery Studio (DS) 2017 Client online tool. RESULTS Among the derivatives, compound 1g2a possessed strong inhibitory activity against HCT116 (IC50 = 5.9 nM) and BEL-7402 (IC50 = 7.8 nM) cells. Compound 1g2a exhibited better selective antiproliferative activities and specificities than all the positive control drugs, including taxol. Compound 1g2a inhibited proliferation of HCT116 and BEL-7402 cells by arresting them in the G2/M phase of the cell cycle, inhibited the migration of HCT116 and BEL-7402 cells and the formation of cell colonies. Compound 1g2a showed excellent tubulin polymerization inhibitory activity on HCT116 and BEL-7402 cells. The results of molecular docking analyses showed that 1g2a may inhibit tubulin to exert anticancer effects. CONCLUSION Compound 1g2a shows outstanding antitumor activity both in vivo and in vitro and has the potential to be further developed into a highly effective antitumor agent with little toxicity to normal tissues.
Collapse
Affiliation(s)
- Ye-Zhi Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Department of Medicinal Chemistry, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, P.R. China
| | - Ya-Bing Xin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Department of Medicinal Chemistry, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, P.R. China
| | - Yuan Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Department of Medicinal Chemistry, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, P.R. China
| | - Xin-Yuan Chen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Department of Medicinal Chemistry, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, P.R. China
| | - De-Ao Man
- Key Laboratory of Natural Medicines of the Changbai Mountain, Department of Medicinal Chemistry, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, P.R. China
| | - Yu-Shun Tian
- Key Laboratory of Natural Medicines of the Changbai Mountain, Department of Medicinal Chemistry, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, P.R. China
| |
Collapse
|
3
|
Ahmed M, Aytacoglu H, Coban O, Tulay P. Investigation of BAK, BAX and MAD2L1 gene expression in human aneuploid blastocysts. ZYGOTE 2023; 31:605-611. [PMID: 37994469 DOI: 10.1017/s0967199423000539] [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] [Indexed: 11/24/2023]
Abstract
Maintaining genomic stability is crucial for normal development. At earlier stages of preimplantation development, as the embryonic genome activation is not fully completed, the embryos may be more prone to abnormalities. Aneuploidies are one of the most common genetic causes of implantation failure or first-trimester miscarriages. Apoptosis is a crucial mechanism to eliminate damaged or abnormal cells from the organism to enable healthy growth. Therefore, this study aimed to determine the relationship between the expression levels of genes involved in apoptosis in human aneuploid and euploid blastocysts. In total, 32 human embryos obtained from 21 patients were used for this study. Trophectoderm biopsies were performed and next-generation screening was carried out for aneuploidy screening. Total RNA was extracted from each blastocyst separately and cDNA was synthesized. Gene expression levels were evaluated using RT-PCR. The statistical analysis was performed to evaluate the gene expression level variations in the euploid and aneuploid embryos, respectively. The expression level of the BAX gene was significantly different between the aneuploid and euploid samples. BAX expression levels were found to be 1.5-fold lower in aneuploid cells. However, the expression levels of BAK and MAD2L1 genes were similar in each group. This study aimed to investigate the possible role of genes involved in apoptosis and aneuploidy mechanisms. The findings of this investigation revealed that the BAX gene was expressed significantly differently between aneuploid and euploid embryos. Therefore, it is possible that the genes involved in the apoptotic pathway have a role in the aneuploidy mechanism.
Collapse
Affiliation(s)
- M Ahmed
- Near East University, Faculty of Medicine, Department of Medical Genetics, Nicosia, Cyprus
| | - H Aytacoglu
- Near East University, Faculty of Medicine, Department of Medical Genetics, Nicosia, Cyprus
| | - O Coban
- British Cyprus IVF Hospital, Embryology Lab, Nicosia, Cyprus
| | - P Tulay
- Near East University, Faculty of Medicine, Department of Medical Genetics, Nicosia, Cyprus
- Near East University, DESAM Research Institute, Nicosia, Cyprus
- Near East University, Center of Excellence, Genetics and Cancer Diagnosis-Research Center, Nicosia, Cyprus
| |
Collapse
|
4
|
Mallardo M, Scalia G, Raia M, Daniele A, Nigro E. The Effects of Adiponectin on the Behavior of B-Cell Leukemia Cells: Insights from an In Vitro Study. Biomedicines 2023; 11:2585. [PMID: 37761026 PMCID: PMC10527421 DOI: 10.3390/biomedicines11092585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Background: Non-Hodgkin's lymphoma (NHL), the most frequent hematological neoplasm worldwide, represents a heterogeneous group of malignancies. The etiology of NHL remains to be fully elucidated, but the role of adipose tissue (AT) in immune function via the secretion of adipokines was recently recognized. Among adipokines, adiponectin has garnered attention for its beneficial properties. This study aimed to explore the in vitro effects of AdipoRon, an adiponectin agonist, on JVM-2, a lymphoblast cell line used as a representative disease model. Methods: JVM-2 cells were treated with different concentrations of AdipoRon to evaluate its effects on viability (via an MTT test), cell cycle distribution (via an FACS analysis), invasiveness (via a Matrigel assay) and colony-forming ability; protein expression was assessed via a real-time PCR (qPCR) and/or Western blotting (WB). Results: We found that the prolonged exposure of JVM-2 cells to AdipoRon led to a reduction in their viability due to a cytostatic effect. Additionally, AdipoRon stimulated both the formation of cell colonies and the expression of E-cadherin. Interestingly, the administration of AdipoRon increased the invasive potential of JVM-2 cells. Conclusions: Our findings indicate that adiponectin is involved in the regulation of different cellular processes of JVM-2 cells, supporting its potential association with a pro-tumorigenic phenotype and indicating that it might contribute to the increased aggressiveness and metastatic potential of B lymphoma cells. However, additional studies are required to fully understand the molecular mechanisms of adiponectin's actions on lymphoblasts and whether it may represent a marker of disease.
Collapse
Affiliation(s)
- Marta Mallardo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche, Farmaceutiche, Università della Campania “Luigi Vanvitelli”, Via A. Vivaldi, 81100 Caserta, Italy; (M.M.); (E.N.)
- CEINGE Biotecnologie Avanzate “Franco Salvatore” Scarl, Via G. Salvatore 486, 80145 Napoli, Italy; (G.S.); (M.R.)
| | - Giulia Scalia
- CEINGE Biotecnologie Avanzate “Franco Salvatore” Scarl, Via G. Salvatore 486, 80145 Napoli, Italy; (G.S.); (M.R.)
| | - Maddalena Raia
- CEINGE Biotecnologie Avanzate “Franco Salvatore” Scarl, Via G. Salvatore 486, 80145 Napoli, Italy; (G.S.); (M.R.)
| | - Aurora Daniele
- CEINGE Biotecnologie Avanzate “Franco Salvatore” Scarl, Via G. Salvatore 486, 80145 Napoli, Italy; (G.S.); (M.R.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Via Pansini, 80131 Napoli, Italy
| | - Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche, Farmaceutiche, Università della Campania “Luigi Vanvitelli”, Via A. Vivaldi, 81100 Caserta, Italy; (M.M.); (E.N.)
- CEINGE Biotecnologie Avanzate “Franco Salvatore” Scarl, Via G. Salvatore 486, 80145 Napoli, Italy; (G.S.); (M.R.)
| |
Collapse
|
5
|
Liu W, Yasui M, Sassa A, You X, Wan J, Cao Y, Xi J, Zhang X, Honma M, Luan Y. FTO regulates the DNA damage response via effects on cell-cycle progression. MUTATION RESEARCH/GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2023; 887:503608. [PMID: 37003652 DOI: 10.1016/j.mrgentox.2023.503608] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/02/2023]
Abstract
The fat mass and obesity-associated protein FTO is an "eraser" of N6-methyladenosine, the most abundant mRNA modification. FTO plays important roles in tumorigenesis. However, its activities have not been fully elucidated and its possible involvement in DNA damage - the early driving event in tumorigenesis - remains poorly characterized. Here, we have investigated the role of FTO in the DNA damage response (DDR) and its underlying mechanisms. We demonstrate that FTO responds to various DNA damage stimuli. FTO is overexpressed in mice following exposure to the promutagens aristolochic acid I and benzo[a]pyrene. Knockout of the FTO gene in TK6 cells, via CRISPR/Cas9, increased genotoxicity induced by DNA damage stimuli (micronucleus and TK mutation assays). Cisplatin- and diepoxybutane-induced micronucleus frequencies and methyl methanesulfonate- and azathioprine-induced TK mutant frequencies were also higher in FTO KO cells. We investigated the potential roles of FTO in DDR. RNA sequencing and enrichment analysis revealed that FTO deletion disrupted the p38 MAPK pathway and inhibited the activation of nucleotide excision repair and cell-cycle-related pathways following cisplatin (DNA intrastrand cross-links) treatment. These effects were confirmed by western blotting and qRT-PCR. FTO deletion impaired cell-cycle arrest at the G2/M phase following cisplatin and diepoxybutane treatment (flow cytometry analysis). Our findings demonstrated that FTO is involved in several aspects of DDR, acting, at least in part, by impairing cell cycle progression.
Collapse
|
6
|
Tan L, Wu C, Zhang J, Yu Q, Wang X, Zhang L, Ge M, Wang Z, Ouyang L, Wang Y. Design, Synthesis, and Biological Evaluation of Heterocyclic-Fused Pyrimidine Chemotypes Guided by X-ray Crystal Structure with Potential Antitumor and Anti-multidrug Resistance Efficacy Targeting the Colchicine Binding Site. J Med Chem 2023; 66:3588-3620. [PMID: 36802449 DOI: 10.1021/acs.jmedchem.2c02115] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Herein, a series of quinazoline and heterocyclic fused pyrimidine analogues were designed and synthesized based on the X-ray co-crystal structure of lead compound 3a, showing efficacious antitumor activities. Two analogues, 15 and 27a, exhibited favorable antiproliferative activities, which were more potent than lead compound 3a by 10-fold in MCF-7 cells. In addition, 15 and 27a exhibited potent antitumor efficacy and tubulin polymerization inhibition in vitro. 15 reduced the average tumor volume by 80.30% (2 mg/kg) in the MCF-7 xenograft model and 75.36% (4 mg/kg) in the A2780/T xenograft model, respectively. Most importantly, supported by structural optimization and Mulliken charge calculation, X-ray co-crystal structures of compounds 15, 27a, and 27b in complex with tubulin were resolved. In summary, our research provided the rational design strategy of colchicine binding site inhibitors (CBSIs) based on X-ray crystallography with antiproliferation, antiangiogenesis, and anti-multidrug resistance properties.
Collapse
Affiliation(s)
- Lun Tan
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chengyong Wu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jifa Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Quanwei Yu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiye Wang
- Department of Pharmacy, Western Theater Command Hospital, Chengdu 610083, Sichuan, China
| | - Lele Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meiyi Ge
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhijia Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Liang Ouyang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
7
|
Hayashi Y, Kimura S, Yano E, Yoshimoto S, Saeki A, Yasukochi A, Hatakeyama Y, Moriyama M, Nakamura S, Jimi E, Kawakubo-Yasukochi T. Id4 modulates salivary gland homeostasis and its expression is downregulated in IgG4-related disease via miR-486-5p. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119404. [PMID: 36535369 DOI: 10.1016/j.bbamcr.2022.119404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/09/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022]
Abstract
Salivary glands are physiologically orchestrated by the coordinated balance between cell differentiation, proliferation, apoptosis, and interactions between epithelial, mesenchymal endothelial, and neuronal cells, and they are frequent sites of manifestations of Sjögren's syndrome (SS) or IgG4-related disease (IgG4-RD). However, little is known about salivary gland homeostasis and its involvement in those diseases. Inhibitor of DNA binding/differentiation 4 (Id4) is an Id protein involved in the transcriptional control of many biological events, including differentiation. Studies of Id4-deficient mice revealed that Id4-deficient submandibular glands were smaller and exhibited accelerated differentiation, compared with those from wild-type littermates. In addition, dry mouth symptoms and Th17 expansion in splenocytes were also observed in the absence of Id4. Furthermore, Id4 levels in the salivary glands of patients with IgG4-RD, but not SS, were significantly decreased compared with those of healthy controls. miRNA-mRNA integrated analysis demonstrated that miR-486-5p was upregulated in IgG4-RD patients and that it might regulate Id4 in the lesion sites. Together, these results provide evidence for the inhibitory role of Id4 in salivary differentiation, and a critical association between Id4 downregulation and IgG4-RD.
Collapse
Affiliation(s)
- Yoshikazu Hayashi
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Soi Kimura
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ena Yano
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shohei Yoshimoto
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka 814-0193, Japan; Oral Medicine Research Center, Fukuoka Dental College, Fukuoka 814-0193, Japan
| | - Ayaka Saeki
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Atsushi Yasukochi
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuji Hatakeyama
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Masafumi Moriyama
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Seiji Nakamura
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Eijiro Jimi
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomoyo Kawakubo-Yasukochi
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
8
|
Deng B, Sun Z, Wang Y, Mai R, Yang Z, Ren Y, Liu J, Huang J, Ma Z, Chen T, Zeng C, Chen J. Design, synthesis, and bioevaluation of imidazo [1,2-a] pyrazine derivatives as tubulin polymerization inhibitors with potent anticancer activities. Bioorg Med Chem 2022; 76:117098. [PMID: 36455508 DOI: 10.1016/j.bmc.2022.117098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
Through structural optimization and ring fusion strategy, we designed a series of novel imidazo[1,2-a]pyrazine derivatives as potential tubulin inhibitors. These compounds displayed potent anti-proliferative activities (micromolar to nanomolar) against a panel of cancer cell lines (including HepG-2, HCT-116, A549 and MDA-MB-231 cells). Among them, compound TB-25 exhibited the strongest inhibitory effects against HCT-116 cells with an IC50 of 23 nM. Mechanism studies revealed that TB-25 could effectively inhibit tubulin polymerization in vitro, and destroy the dynamic equilibrium of microtubules in HCT-116 cells. In addition, TB-25 dose-dependently induced G2/M phase cell cycle arrest and apoptosis in HCT-116 cells. Furthermore, TB-25 suppressed HCT-116 cell migration in a concentration-dependent manner. Finally, molecular docking showed that TB-25 fitted well in the colchicine binding site of tubulin and overlapped nicely with CA-4. Collectively, these results suggest that TB-25 represents a promising tubulin inhibitor deserving further investigation.
Collapse
Affiliation(s)
- Bulian Deng
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Zhiqiang Sun
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Precision Medicine Research Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruiyao Mai
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Zichao Yang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Yichang Ren
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Jin Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Junli Huang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Zeli Ma
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ting Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Canjun Zeng
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
9
|
Seymore TN, Rivera-Núñez Z, Stapleton PA, Adibi JJ, Barrett ES. Phthalate Exposures and Placental Health in Animal Models and Humans: A Systematic Review. Toxicol Sci 2022; 188:153-179. [PMID: 35686923 PMCID: PMC9333406 DOI: 10.1093/toxsci/kfac060] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Phthalates are ubiquitous compounds known to leach from the plastic products that contain them. Due to their endocrine-disrupting properties, a wide range of studies have elucidated their effects on reproduction, metabolism, neurodevelopment, and growth. Additionally, their impacts during pregnancy and on the developing fetus have been extensively studied. Most recently, there has been interest in the impacts of phthalates on the placenta, a transient major endocrine organ critical to maintenance of the uterine environment and fetal development. Phthalate-induced changes in placental structure and function may have significant impacts on the course of pregnancy and ultimately, child health. Prior reviews have described the literature on phthalates and placental health; however to date, there has been no comprehensive, systematic review on this topic. Here, we review 35 papers (24 human and 11 animal studies) and summarize phthalate exposures in relation to an extensive set of placental measures. Phthalate-related alterations were reported for placental morphology, hormone production, vascularization, histopathology, and gene/protein expression. The most consistent changes were observed in vascular and morphologic endpoints, including cell composition. These changes have implications for pregnancy complications such as preterm birth and intrauterine growth restriction as well as potential ramifications for children's health. This comprehensive review of the literature, including common sources of bias, will inform the future work in this rapidly expanding field.
Collapse
Affiliation(s)
- Talia N Seymore
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey 08854, USA
| | - Zorimar Rivera-Núñez
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey 08854, USA
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey 08854, USA
| | - Phoebe A Stapleton
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey 08854, USA
| | - Jennifer J Adibi
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | - Emily S Barrett
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey 08854, USA
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey 08854, USA
| |
Collapse
|
10
|
Wang ZZ, Meng T, Yang MY, Wang W, Zhang Y, Liu Y, Han AQ, Wu J, Wang HX, Qian B, Zhu LX. ALYREF associated with immune infiltration is a prognostic biomarker in hepatocellular carcinoma. Transl Oncol 2022; 21:101441. [PMID: 35523010 PMCID: PMC9079359 DOI: 10.1016/j.tranon.2022.101441] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/07/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022] Open
Abstract
ALYREF is a potential prognostic marker for hepatocellular carcinoma. ALYREF affects the biological function of hepatocellular carcinoma cells. ALYREF is associated with immune infiltration in hepatocellular carcinoma. The model constructed based on ALYREF-related immune genes provides a reference for the evaluation of immunotherapy.
Background Although ALYREF has been demonstrated to have a role in a number of malignancies, its role in hepatocellular carcinoma (HCC) has received little attention. Our objective was to research at the prognostic value, biological role and relevance of ALYREF to the immune system in HCC. Methods The expression of ALYREF and its relationship with clinical parameters of HCC patients were analyzed by liver cancer cohort (LIHC) of The Cancer Genome Atlas. The expression and prognosis were verified by immunohistochemistry experiments. Gene transfection, CCK-8, scratch healing, transwell invasion and flow cytometry were used to assess the molecular function of ALYREF in vitro. The TIMER and TISIDB online data portals were used to assess the relevance of ALYREF to immunization. Stepwise regression analysis of ALYREF-related immune genes in the LIHC training set was used to construct a prognostic risk prediction model. Also, construct a nomogram to predict patient survival. The testing set for internal verification. Results Knockdown of ALYREF changed the biological phenotypes of HCC cells, such as proliferation, apoptosis, and invasion. In addition, the expression of ALYREF in HCC affected the level of immune cell infiltration and correlated with the overall survival time of patients. The constructed immune prognostic model allows for a valid assessment of patients. Conclusion ALYREF is increased in HCC, has an impact on cellular function and the immune system, and might be used as a prognostic marker.
Collapse
Affiliation(s)
- Zhen-Zhen Wang
- Department of General Surgery, Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Tao Meng
- Department of General Surgery, The First People's Hospital of Hefei, Hefei 230000, China
| | - Ming-Ya Yang
- Department of Haematology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Wei Wang
- Department of General Surgery, Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yan Zhang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yu Liu
- Department of General Surgery, Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - An-Qi Han
- Department of General Surgery, Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Jin Wu
- Department of General Surgery, Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Hui-Xiao Wang
- Department of Medicine, The Second People's Hospital of Anhui Province, Hefei 230000, China.
| | - Bo Qian
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Li-Xin Zhu
- Department of General Surgery, Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
11
|
Sun CC, Zhao S, Chu LL, Zhang SY, Li YL, Sun MF, Wang QN, Huang Y, Zhang J, Wang H, Gao L, Xu DX, Zhang SC, Xu T, Zhao LL. Di (2-ethyl-hexyl) phthalate disrupts placental growth in a dual blocking mode. JOURNAL OF HAZARDOUS MATERIALS 2022; 421:126815. [PMID: 34396966 DOI: 10.1016/j.jhazmat.2021.126815] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 06/13/2023]
Abstract
Di (2-ethyl-hexyl) phthalate (DEHP) is a widely used plasticizer. Maternal DEHP exposure inhibits cell proliferation and reduces placentas size, which associates with fetal growth restriction and adulthood diseases. However, the mechanism of placental cell proliferation inhibition by DEHP remains elusive. This study investigated the effect of DEHP on placental cell proliferation from cell cycle arrest. Utilizing in vitro and in vivo experiments, we investigated cell cycle arrest, DNA double-strand break (DSB) repair, genotoxic stress response, and micronuclei formation. Most DEHP metabolizes to mono (2-Ethylhexyl) phthalate (MEHP) and distributes to organs quickly, so MEHP and DEHP were used in cultured cell and animal experiments, respectively. Here, a double blocking mode for the proliferation inhibition of the placental cell was revealed. One is that the classical DSB repair pathways were suppressed, which arrested the cell cycle at the G2/M phase. The other is that DEHP stimulated an elevated level of progesterone, which blocked the cell cycle at metaphase by disrupting chromosome arrangement. These two sets of events facilitated micronuclei formation and resulted in cell proliferation inhibition. This findings provide a novel mechanistic understanding for DEHP to inhibit placental cell proliferation.
Collapse
Affiliation(s)
- Cong-Cong Sun
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; School of Biology, Food and Environment, Hefei University, Hefei 230601, China; Tongxiang Centre for Disease Control and Prevention, Tongxiang 314500, Zhejiang Province, China
| | - Shuai Zhao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; School of Biology, Food and Environment, Hefei University, Hefei 230601, China
| | - Ling-Luo Chu
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Shan-Yu Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; School of Biology, Food and Environment, Hefei University, Hefei 230601, China
| | - Yan-Ling Li
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Mei-Fang Sun
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Qu-Nan Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Yichao Huang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Jun Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Hua Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Lan Gao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - De-Xiang Xu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Shi-Chen Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Tao Xu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; School of Biology, Food and Environment, Hefei University, Hefei 230601, China.
| | - Ling-Li Zhao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Department of Toxicology/Anhui Provincial Key Laboratory of Population Health and Aristogenics/MOE Key Laboratory of Population Health Across Life Cycle, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China.
| |
Collapse
|
12
|
Liu J, Hao Y, He Y, Li X, Sun DE, Zhang Y, Yang PY, Chen X. Quantitative and Site-Specific Chemoproteomic Profiling of Protein O-GlcNAcylation in the Cell Cycle. ACS Chem Biol 2021; 16:1917-1923. [PMID: 34161081 DOI: 10.1021/acschembio.1c00301] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mammalian cell cycle is a central process for tissue growth and maintenance. Protein O-linked β-N-acetylglucosamine (O-GlcNAc) modification has been found to occur on several important cell cycle regulators. However, the O-GlcNAcylated proteome has not been extensively profiled during cell cycle progression. Herein, we report a quantitative profiling of protein O-GlcNAcylation sites in cell proliferation, by using an O-GlcNAc chemoproteomic strategy. In HeLa cells, a total of 902, 439, and 872 high-confidence O-GlcNAcylation sites distributed on 414, 265, and 425 proteins are identified in the interphase, early mitosis, and mitotic exit stages, respectively. The identified O-GlcNAcylation events occur on a variety of important regulators, which are involved in the processes of cell division, DNA repair, and cell death. Furthermore, we show that O-GlcNAcylation is dynamically regulated in a cell cycle stage-dependent manner. Our results provide a valuable resource for investigating the functional roles of O-GlcNAc in the mammalian cell cycle.
Collapse
Affiliation(s)
- Jialin Liu
- Institute of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Yi Hao
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - Yanwen He
- College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Xiang Li
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - De-en Sun
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
| | - Yang Zhang
- Institute of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Peng-Yuan Yang
- Institute of Biomedical Sciences, Fudan University, Shanghai 200433, China
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Synthetic and Functional Biomolecules Center, Peking University, Beijing 100871, China
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
| |
Collapse
|
13
|
Kinases leave their mark on caspase substrates. Biochem J 2021; 478:3179-3184. [PMID: 34492095 DOI: 10.1042/bcj20210399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022]
Abstract
Apoptosis is a cell death program that is executed by the caspases, a family of cysteine proteases that typically cleave after aspartate residues during a proteolytic cascade that systematically dismantles the dying cell. Extensive signaling crosstalk occurs between caspase-mediated proteolysis and kinase-mediated phosphorylation, enabling integration of signals from multiple pathways into the decision to commit to apoptosis. A new study from Maluch et al. examines how phosphorylation within caspase cleavage sites impacts the efficiency of substrate cleavage. The results demonstrate that while phosphorylation in close proximity to the scissile bond is generally inhibitory, it does not necessarily abrogate substrate cleavage, but instead attenuates the rate. In some cases, this inhibition can be overcome by additional favorable substrate features. These findings suggest potential nuanced physiological roles for phosphorylation of caspase substrates with exciting implications for targeting caspases with chemical probes and therapeutics.
Collapse
|
14
|
Wang K, Zhong H, Li N, Yu N, Wang Y, Chen L, Sun J. Discovery of Novel Anti-Breast-Cancer Inhibitors by Synergistically Antagonizing Microtubule Polymerization and Aryl Hydrocarbon Receptor Expression. J Med Chem 2021; 64:12964-12977. [PMID: 34428056 DOI: 10.1021/acs.jmedchem.1c01099] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A series of unreported dual-receptor inhibitors targeting both the tubulin colchicine site and AhR were designed and synthesized, and their anti-breast-cancer activities were evaluated. Compound 12 showed the strongest activity with an IC50 of 0.9 nM in MCF-7 cell lines. Besides, 12 could significantly inhibit cancer growth in MCF-7 xenograft tumor models with no obvious toxic effects and was more effective than the positive control (combretastatin A-4). With the in-depth study, it was found that 12 could induce apoptosis in breast cancer cells by making arrest in G2/M phase, depolarizing mitochondria and inducing intracellular reactive oxygen generation. This evident anticancer effect and the ability to inhibit cell migration were attributed to the synergistic antagonism of 12 on tubulin and AhR. In general, 12 was worthy of further research as an effective and safe anti-breast-cancer drug.
Collapse
Affiliation(s)
- Kun Wang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Hui Zhong
- Department of Pharmacology of Traditional Chinese Medicine, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 210009, China
| | - Na Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Nairong Yu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yujin Wang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Li Chen
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Jianbo Sun
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| |
Collapse
|
15
|
Banerjee S, Mahmud F, Deng S, Ma L, Yun MK, Fakayode SO, Arnst KE, Yang L, Chen H, Wu Z, Lukka PB, Parmar K, Meibohm B, White SW, Wang Y, Li W, Miller DD. X-ray Crystallography-Guided Design, Antitumor Efficacy, and QSAR Analysis of Metabolically Stable Cyclopenta-Pyrimidinyl Dihydroquinoxalinone as a Potent Tubulin Polymerization Inhibitor. J Med Chem 2021; 64:13072-13095. [PMID: 34406768 PMCID: PMC9206499 DOI: 10.1021/acs.jmedchem.1c01202] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Small molecules that interact with the colchicine binding site in tubulin have demonstrated therapeutic efficacy in treating cancers. We report the design, syntheses, and antitumor efficacies of new analogues of pyridopyrimidine and hydroquinoxalinone compounds with improved drug-like characteristics. Eight analogues, 5j, 5k, 5l, 5m, 5n, 5r, 5t, and 5u, showed significant improvement in metabolic stability and demonstrated strong antiproliferative potency in a panel of human cancer cell lines, including melanoma, lung cancer, and breast cancer. We report crystal structures of tubulin in complex with five representative compounds, 5j, 5k, 5l, 5m, and 5t, providing direct confirmation for their binding to the colchicine site in tubulin. A quantitative structure-activity relationship analysis of the synthesized analogues showed strong ability to predict potency. In vivo, 5m (4 mg/kg) and 5t (5 mg/kg) significantly inhibited tumor growth as well as melanoma spontaneous metastasis into the lung and liver against a highly paclitaxel-resistant A375/TxR xenograft model.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Physical Sciences, College of STEM, University of Arkansas Fort Smith, Fort Smith, Arkansas 72913, United States
| | - Foyez Mahmud
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Lingling Ma
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mi-Kyung Yun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Sayo O Fakayode
- Department of Physical Sciences, College of STEM, University of Arkansas Fort Smith, Fort Smith, Arkansas 72913, United States
| | - Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Zhongzhi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Pradeep B Lukka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Keyur Parmar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Stephen W White
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
16
|
Ang KP, Chan PF, Hamid RA. Induction of apoptosis on ovarian adenocarcinoma cells, A2780 by tricyclohexylphosphanegold (I) mercaptobenzoate derivatives via intrinsic and extrinsic pathways. J Biol Inorg Chem 2021; 26:833-853. [PMID: 34476610 DOI: 10.1007/s00775-021-01892-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/17/2021] [Indexed: 01/10/2023]
Abstract
Tricyclohexylphosphanegold(I) n-mercaptobenzoate (n = 2, 3, 4) labelled as 1-3 were previously reported to significantly suppress thioredoxin reductase (TrxR) activities towards ovarian cancer cells, A2780, in vitro. Herein, we explored the role of 1-3 for their apoptosis inducing ability against A2780 cells. 1-3 exhibited IC50 values at 1.19 ± 0.03 µM, 2.28 ± 0.04 μM and 0.78 ± 0.01 μM, respectively, compared to cisplatin at 26.8 ± 0.15 µM. The compounds induced A2780 apoptosis via a caspase-dependent mitochondrion pathway as evidenced by ROS production, cytochrome c release, caspases-3/7, -8, -9 and -10 activation, APAF1 and BAX upregulation as well as BCL2A1 and BCL2 genes' downregulation. In addition, the death mode of 1-3 was also mediated via death receptor extrinsic pathway manifested by FAS, FASL, FADD, and TNFR1 genes' upregulation via Human Rt PCR analysis. In addition, 1-3 significantly caused A2780 arrest at S phase, which was associated with the upregulation of TP53, E2F1, RB1 and CDKN1A upregulation and downregulation of CDK1, CDK4, CDC25A and CDC25C genes. Based on these promising results, these phosphanegold(I) thiolate derivatives could act as feasible candidates for further advanced in vivo ovarian cancer studies to develop novel chemotherapeutic agents derived from metal-based agents.
Collapse
Affiliation(s)
- Kok Pian Ang
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Pit Foong Chan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Roslida Abd Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
17
|
Wu CJ, Wu JQ, Hu Y, Pu S, Lin Y, Zeng Z, Hu J, Chen WH. Design, synthesis and biological evaluation of indole-based [1,2,4]triazolo[4,3-a] pyridine derivatives as novel microtubule polymerization inhibitors. Eur J Med Chem 2021; 223:113629. [PMID: 34175541 DOI: 10.1016/j.ejmech.2021.113629] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/23/2021] [Accepted: 06/08/2021] [Indexed: 11/29/2022]
Abstract
A series of indole-based [1,2,4]triazolo [4,3-a]pyridine derivatives was designed and synthesized as novel microtubulin polymerization inhibitors by using a conformational restriction strategy. These compounds exhibited moderate to potent anti-proliferative activities against a panel of cancer cell lines (HeLa, A549, MCF-7 and HCT116). Among them, compound 12d featuring a N-methyl-5-indolyl substituent at the C-6 position of the [1,2,4]triazolo [4,3-a]pyridine core exhibited the highest antiproliferative activity with the IC50 values ranging from 15 to 69 nM, and remarkable inhibitory effect on tubulin polymerization with an IC50 value of 1.64 μM. Mechanistic studies revealed that compound 12d induced cellular apoptosis and cell cycle arrest at the G2/M phase in a dose-dependent fashion. Moreover, compound 12d significantly suppressed wound closure and disturbed microtubule networks.
Collapse
Affiliation(s)
- Cheng-Jun Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Jia-Qiang Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Yunfei Hu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Suyun Pu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Yuying Lin
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Zimai Zeng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Jinhui Hu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China.
| | - Wen-Hua Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China.
| |
Collapse
|
18
|
Sun M, Zhang Y, Qin J, Ba M, Yao Y, Duan Y, Liu H, Yu D. Synthesis and biological evaluation of new 2-methoxyestradiol derivatives: Potent inhibitors of angiogenesis and tubulin polymerization. Bioorg Chem 2021; 113:104988. [PMID: 34034135 DOI: 10.1016/j.bioorg.2021.104988] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022]
Abstract
Here, we report the structural optimization of a hit natural compound, 2-ME2 (2-methoxyestradiol), which exhibited inhibitory activity but low potency on tubulin polymerization, anti- angiogenesis, MCF-7 proliferation and metastasis in vitro and in vivo. A novel series of 3,17-modified and 17-modified analogs of 2-ME2 were synthesized and investigated for their antiproliferative activity against MCF-7 and another five different human cancer cell lines leading to the discovery of 9i. 9i bind to tubulin colchicine site tightly, inhibited tubulin polymerization and disrupted cellular microtubule networks. Cellular mechanism studies revealed that 9i could induce G2/M phase arrest by down-regulated expression of p-Cdc2, P21 and cell apoptosis by regulating apoptosis-related proteins (Parp, Caspase families) in a dose-dependent manner. Importantly, 9i significantly inhibited HUVEC tube formation, proliferation, migration and invasion. The inhibitory effect against angiogenesis in vivo was confirmed by zebrafish xenograft. Furthermore, 9i could effectively inhibit the proliferation and metastasis of MCF-7 cells in vitro and in zebrafish xenograft. The satisfactory physicochemical property and metabolic stability of 9i further indicated that it can act as a promising and potent anti-angiogenesis, inhibiting proliferation and metastasis of breast cancer agent via targeting tubulin colchicine binding site.
Collapse
Affiliation(s)
- Moran Sun
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yixin Zhang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Jinling Qin
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Mengyu Ba
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yongfang Yao
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China.
| | - Hongmin Liu
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Dequan Yu
- Chinese Academy of Medical Sciences, Beijing 100021,China
| |
Collapse
|
19
|
Hao SY, Qi ZY, Wang S, Wang XR, Chen SW. Synthesis and bioevaluation of N-(3,4,5-trimethoxyphenyl)-1H-pyrazolo[3,4-b]pyridin-3-amines as tubulin polymerization inhibitors with anti-angiogenic effects. Bioorg Med Chem 2021; 31:115985. [DOI: 10.1016/j.bmc.2020.115985] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 02/08/2023]
|
20
|
Development of triazolothiadiazine derivatives as highly potent tubulin polymerization inhibitors: Structure-activity relationship, in vitro and in vivo study. Eur J Med Chem 2020; 208:112847. [DOI: 10.1016/j.ejmech.2020.112847] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022]
|
21
|
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: 9.0] [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
|
22
|
Yang F, Jian XE, Diao PC, Huo XS, You WW, Zhao PL. Synthesis, and biological evaluation of 3,6-diaryl-[1,2,4]triazolo[4,3-a]pyridine analogues as new potent tubulin polymerization inhibitors. Eur J Med Chem 2020; 204:112625. [DOI: 10.1016/j.ejmech.2020.112625] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/08/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023]
|
23
|
New Paracyclophanylthiazoles with Anti-Leukemia Activity: Design, Synthesis, Molecular Docking, and Mechanistic Studies. Molecules 2020; 25:molecules25133089. [PMID: 32645912 PMCID: PMC7411887 DOI: 10.3390/molecules25133089] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 11/23/2022] Open
Abstract
A new series of methyl 2-(2-(4′-[2.2]paracyclophanyl)-hydrazinylidene)-3-substituted-4-oxothiazolidin-5-ylidene)acetates 3a–f were synthesized from the reaction of paracyclophanyl-acylthiosemicarbazides 2a–f with dimethyl acetylenedicarboxylate. Based upon nuclear magnetic resonance (NMR), infrared (IR), and mass spectra (HRMS), the structure of the obtained products was elucidated. X-ray structure analysis was also used as unambiguous tool to elucidate the structure of the products. The target compounds 3a–f were screened against 60 cancer cell lines. They displayed anticancer activity against a leukemia subpanel, namely, RPMI-8226 and SR cell lines. The activity of compound 3a was found as the most cytotoxic potency against 60 cancer cell lines. Consequently, it was selected for further five doses analysis according to National Cancer Institute (NCI) protocol. The cytotoxic effect showed selectivity ratios ranging between 0.63 and 1.28 and between 0.58 and 5.89 at the GI50 and total growth inhibition (TGI) levels, respectively. Accordingly, compound 3a underwent further mechanistic study against the most sensitive leukemia RPMI-8226 and SR cell lines. It showed antiproliferation with IC50 = 1.61 ± 0.04 and 1.11 ± 0.03 µM against RPMI-8226 and SR cell lines, respectively. It also revealed a remarkable tubulin inhibitory activity, compared to colchicine with IC50 = 4.97 µM/mL. Caspase-3, BAX, and Bcl-2 assays for 3a using annexin V-FITC staining revealed significant pro-apoptotic activity. Furthermore, multidrug-resistant leukemia SR cells were used to show better resistance indices (1.285 ng/mL, 1.15-fold) than the reference. Docking studies with β-tubulin indicate that most of the tested compounds illustrated good binding at the colchicine binding site of the enzyme, especially for compound 3a, which made several interactions better than that of the reference colchicine.
Collapse
|
24
|
Abstract
The goal of mitosis is to form two daughter cells each containing one copy of each mother cell chromosome, replicated in the previous S phase. To achieve this, sister chromatids held together back-to-back at their primary constriction, the centromere, have to interact with microtubules of the mitotic spindle so that each chromatid takes connections with microtubules emanating from opposite spindle poles (we will refer to this condition as bipolar attachment). Only once all replicated chromosomes have reached bipolar attachments can sister chromatids lose cohesion with each other, at the onset of anaphase, and move toward opposite spindle poles, being segregated into what will soon become the daughter cell nucleus. Prevention of errors in chromosome segregation is granted by a safeguard mechanism called Spindle Assembly Checkpoint (SAC). Until all chromosomes are bipolarly oriented at the equator of the mitotic spindle, the SAC prevents loss of sister chromatid cohesion, thus anaphase onset, and maintains the mitotic state by inhibiting inactivation of the major M phase promoting kinase, the cyclin B-cdk1 complex (Cdk1). Here, we review recent mechanistic insights about the circuitry that links Cdk1 to the SAC to ensure correct achievement of the goal of mitosis.
Collapse
Affiliation(s)
- Angela Flavia Serpico
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy.,DMMBM, University of Naples "Federico II", Naples, 80131, Italy
| | - Domenico Grieco
- CEINGE Biotecnologie Avanzate, Naples, 80145, Italy.,Department of Pharmacy, University of Naples "Federico II", Naples, 80131, Italy
| |
Collapse
|
25
|
Hua S, Chen F, Wang X, Gou S. Dual-functional conjugates improving cancer immunochemotherapy by inhibiting tubulin polymerization and indoleamine-2,3-dioxygenase. Eur J Med Chem 2020; 189:112041. [PMID: 31954880 DOI: 10.1016/j.ejmech.2020.112041] [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: 09/26/2019] [Revised: 01/05/2020] [Accepted: 01/06/2020] [Indexed: 12/21/2022]
Abstract
A series of novel conjugates comprising tublin and IDO inhibitors were designed, synthesized and evaluated for their antiproliferative activity. Among them, HI5, composed of combretastatin A-4 (CA-4) and (D)-1-methyltryptophan (D-MT) by a linker, exhibited the most potent antitumor activity, in particular with higher IC50 value (0.07 μM) than CA-4 (0.21 μM) against HeLa cancer cell line. Mechanism studies indicated that HI5 can inhibit tubulin polymerization and cell migration, cause G2/M phase arrest, concurrent induce apoptosis via the mitochondrial dependent apoptosis pathway and cause reactive oxidative stress generation in HeLa cells. Furthermore, HI5 can inhibit IDO expression and decrease kynurenine production, leading to stimulating T cells activation and proliferation to enhance antitumor immunity in vitro. Interestingly, HI5 can effectively limit the tumor growth in the HeLa xenograft mice models without causing significant loss of body weight. Consequently, such a conjugation can be a potent and safe immunochemotherapeutic method for improving cancer therapy.
Collapse
Affiliation(s)
- Shixian Hua
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Nanjing 211189, China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Feihong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Nanjing 211189, China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Xinyi Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Nanjing 211189, China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Nanjing 211189, China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China.
| |
Collapse
|
26
|
Zhao X, Liu Z, Shen J, Yong L, Xia Y, Bian M. microRNA-196a Overexpression Inhibits Apoptosis in Hemin-Induced K562 Cells. DNA Cell Biol 2020; 39:235-243. [PMID: 31913716 DOI: 10.1089/dna.2019.5061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
microRNAs (miRNAs) have a crucial role in erythropoiesis. However, the understanding of the apoptosis of erythroid lineage remains poorly understood. Hence, an additional examination is required. K562 cell lines can be differentiated into early erythrocytes by hemin and the model of early erythrocytes can be established, consequently. miR-196a has been proven to take part in antiapoptosis in many cell lines. However, the role of miR-196a associated with the apoptosis in hemin-induced K562 cells remains unclear. To study the potential function of miR-196a involved in the common progenitor of erythroblasts, miR-196a mimics and microRNA-small hairpin negative control (miRNA-ShNC) were transfected into hemin-induced K562 cells with lentiviruses. After that, the viability of the transfected hemin-induced K562 cells was tested by CCK-8 assay, and the alteration of cell cycle and apoptosis rate were detected by flow cytometry. Furthermore, bioinformatics and dual-luciferase report system verified that p27kip1 is a target gene of miR-196a. Additionally, the expression of some proteins associated with cell cycle and apoptosis was tested by Western blotting assays. It was found that after overexpressing miR-196a, the proliferation of hemin-induced K562 cells was promoted while the apoptosis inhibited. Furthermore, miR-196a combines with the 3'UTR of p27kip1 directly. Additionally, the relationship between miR-196a and the protein level of p27kip1 is negative. After restoring the expression of p27kip1, the growth rate of hemin-induced K562 cells was not as high as before and the inhibition of apoptosis was alleviated. The present study validates that miR-196a overexpression inhibits apoptosis in hemin-induced K562 cells through downregulating p27kip1.
Collapse
Affiliation(s)
- Xingyun Zhao
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhenfei Liu
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jijia Shen
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Hefei, China
| | - Liang Yong
- Institute and Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanyuan Xia
- Medical Genetics Center, Anhui Women and Child Health Care Hospital, Hefei, China
| | - Maohong Bian
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Pocasap P, Weerapreeyakul N, Thumanu K. Alyssin and Iberin in Cruciferous Vegetables Exert Anticancer Activity in HepG2 by Increasing Intracellular Reactive Oxygen Species and Tubulin Depolymerization. Biomol Ther (Seoul) 2019; 27:540-552. [PMID: 31405267 PMCID: PMC6824623 DOI: 10.4062/biomolther.2019.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/19/2019] [Accepted: 06/17/2019] [Indexed: 11/23/2022] Open
Abstract
To determine the chemopreventive potential of alyssin and iberin, the in vitro anticancer activities and molecular targets of isothiocyanates (ITCs) were measured and compared to sulforaphane in hepatocellular carcinoma cell HepG2. The SR-FTIR spectra observed a similar pattern vis-à-vis the biomolecular alteration amongst the ITCs-treated cells suggesting a similar mode of action. All of the ITCs in this study cause cancer cell death through both apoptosis and necrosis in concentration dependent manner (20–80 μM). We found no interactions of any of the ITCs studied with DNA. Notwithstanding, all of the ITCs studied increased intracellular reactive oxygen species (ROS) and suppressed tubulin polymerization, which led to cell-cycle arrest in the S and G2/M phase. Alyssin possessed the most potent anticancer ability; possibly due to its ability to increase intracellular ROS rather than tubulin depolymerization. Nevertheless, the structural influence of alkyl chain length on anticancer capabilities of ITCs remains inconclusive. The results of this study indicate an optional, potent ITC (viz., alyssin) because of its underlying mechanisms against hepatic cancer. As a consequence, further selection and development of effective chemotherapeutic ITCs is recommended.
Collapse
Affiliation(s)
- Piman Pocasap
- Research and Development in Pharmaceuticals Program, Graduate School, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand.,Human High Performance and Health Promotion Research Institute (HHP&HP), Khon Kaen University, Khon Kaen 40002, Thailand
| | - Kanjana Thumanu
- Synchrotron Light Research Institute (Public Organization), Nakhon Ratchasima 30000, Thailand
| |
Collapse
|
28
|
Sawada JI, Ishii H, Matsuno K, Sato M, Suzuki Y, Asai A. Selective Inhibition of Spindle Microtubules by a Tubulin-Binding Quinazoline Derivative. Mol Pharmacol 2019; 96:609-618. [PMID: 31471455 DOI: 10.1124/mol.119.116624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/20/2019] [Indexed: 02/03/2023] Open
Abstract
In the research field of tubulin-binding agents for the development of anticancer agents, hidden targets are emerging as a problem in understanding the exact mechanisms of actions. The quinazoline derivative 1-(4-methoxyphenyl)-1-(quinazolin-4-yl)ethan-1-ol (PVHD121) has anti-cell proliferative activity and inhibits tubulin polymerization by binding to the colchicine site of tubulin. However, the molecular mechanism of action of PVHD121 in cells remains unclear. Here, we demonstrate that PVHD121 delays mitotic entry and efficiently causes mitotic arrest with spindle checkpoint activation, leading to subsequent cell death. The dominant phenotype induced by PVHD121 was aberrant spindles with robust microtubules and unseparated centrosomes. The microtubules were radially distributed, and their ends appeared to adhere to kinetochores, and not to centrosomes. Extensive inhibition by high concentrations of PVHD121 eliminated all microtubules from cells. PVHD277 [1-(4-methoxyphenyl)-1-(2-morpholinoquinazolin-4-yl)ethan-1-ol], a PVHD121 derivative with fluorescence, tended to localize close to the centrosomes when cells prepared to enter mitosis. Our results show that PVHD121 is an antimitotic agent that selectively disturbs microtubule formation at centrosomes during mitosis. This antimitotic activity can be attributed to the targeting of centrosome maturation in addition to the interference with microtubule dynamics. Due to its unique bioactivity, PVHD121 is a potential tool for studying the molecular biology of mitosis and a potential lead compound for the development of anticancer agents. SIGNIFICANCE STATEMENT: Many tubulin-binding agents have been developed as potential anticancer agents. The aim of this study was to understand the subcellular molecular actions of a quinazoline derivative tubulin-binding agent, 1-(4-methoxyphenyl)-1-(quinazolin-4-yl)ethan-1-ol (PVHD121). As expected from its binding activity to tubulin, PVHD121 caused aberrant spindles and inhibited mitotic progression. However, in addition to tubulin, PVHD121 also targeted an unexpected biomolecule involved in centrosome maturation. Due to targeting the biomolecule just before entering mitosis, PVHD121 preferentially inhibited centrosome-derived microtubules rather than chromosome-derived microtubules during spindle formation. This study not only revealed the molecular action of PVHD121 in cells but also emphasized the importance of considering possible tubulin-independent effects of tubulin-binding agents via hidden targeted biomolecules for future use.
Collapse
Affiliation(s)
- Jun-Ichi Sawada
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences (J.-i.S., H.I., K.M., A.A.) and Laboratory of Organic Chemistry, School of Pharmaceutical Sciences (M.S., Y.S.), University of Shizuoka, Shizuoka, Japan
| | - Hirosuke Ishii
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences (J.-i.S., H.I., K.M., A.A.) and Laboratory of Organic Chemistry, School of Pharmaceutical Sciences (M.S., Y.S.), University of Shizuoka, Shizuoka, Japan
| | - Kenji Matsuno
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences (J.-i.S., H.I., K.M., A.A.) and Laboratory of Organic Chemistry, School of Pharmaceutical Sciences (M.S., Y.S.), University of Shizuoka, Shizuoka, Japan
| | - Masayuki Sato
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences (J.-i.S., H.I., K.M., A.A.) and Laboratory of Organic Chemistry, School of Pharmaceutical Sciences (M.S., Y.S.), University of Shizuoka, Shizuoka, Japan
| | - Yumiko Suzuki
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences (J.-i.S., H.I., K.M., A.A.) and Laboratory of Organic Chemistry, School of Pharmaceutical Sciences (M.S., Y.S.), University of Shizuoka, Shizuoka, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences (J.-i.S., H.I., K.M., A.A.) and Laboratory of Organic Chemistry, School of Pharmaceutical Sciences (M.S., Y.S.), University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
29
|
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: 3.4] [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
|
30
|
Antitumor activity of methyl (Z)-2-(isothioureidomethyl)-2-pentenoate hydrobromide against leukemia cell lines via mitotic arrest and apoptotic pathways. Biochim Biophys Acta Gen Subj 2019; 1863:1332-1342. [PMID: 31170497 DOI: 10.1016/j.bbagen.2019.05.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 01/07/2023]
Abstract
In a previous study, we described a series of 28 aryl- and alkyl-substituted isothiouronium salts with antitumor activity and selectivity toward a leukemia cell line. Among the synthesized compounds, methyl (Z)-2-(isothioureidomethyl)-2-pentenoate hydrobromide (IS-MF08) showed conspicuous activity. In the present study, we investigated the mechanism of action of IS-MF08. Our results showed that its mechanism most likely is related with the membrane receptor Fas and subsequent activation of the extrinsic cell death pathway, triggered by a decrease in the levels of the anti-apoptotic protein Bcl-2 and caspase-8 and -3 cascade activation, causing DNA damage and mitotic arrest. IS-MF08 also caused an increase in intracellular ROS, endoplasmic reticulum (ER) stress, and mitochondrial membrane permeabilization, resulting in organelle degradation as an attempt to reestablish cell homeostasis. Furthermore, cells exposed to IS-MF08 combined to an autophagy inhibitor were less susceptible to compound's cytotoxicity, suggesting that autophagy makes part of its mechanism of action. These data support the hypothesis that IS-MF08 acts by the apoptosis extrinsic pathway and possibly by autophagy as mechanisms of cell death.
Collapse
|
31
|
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: 1.8] [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
|
32
|
Mishra SK, Stephenson DJ, Chalfant CE, Brown RE. Upregulation of human glycolipid transfer protein (GLTP) induces necroptosis in colon carcinoma cells. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:158-167. [PMID: 30472325 PMCID: PMC6448591 DOI: 10.1016/j.bbalip.2018.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/03/2018] [Accepted: 11/10/2018] [Indexed: 02/06/2023]
Abstract
Human GLTP on chromosome 12 (locus 12q24.11) encodes a 24 kD amphitropic lipid transfer protein (GLTP) that mediates glycosphingolipid (GSL) intermembrane trafficking and regulates GSL homeostatic levels within cells. Herein, we provide evidence that GLTP overexpression inhibits the growth of human colon carcinoma cells (HT-29; HCT-116), but spares normal colonic cells (CCD-18Co). Mechanistic studies reveal that GLTP overexpression arrested the cell cycle at the G1/S checkpoint via upregulation of cyclin-dependent kinase inhibitor-1B (Kip1/p27) and cyclin-dependent kinase inhibitor 1A (Cip1/p21) at the protein and mRNA levels, and downregulation of cyclin-dependent kinase-2 (CDK2), cyclin-dependent kinase-4 (CDK4), cyclin E and cyclin D1 protein levels. Assessment of the biological fate of HCT-116 cells overexpressing GLTP indicated no increase in cell death suggesting induction of quiescence. However, HT-29 cells overexpressing GLTP underwent cell death by necroptosis as revealed by phosphorylation of human mixed lineage kinase domain-like protein (pMLKL) via receptor-interacting protein kinase-3 (RIPK-3), elevated cytosolic calcium, and plasma membrane permeabilization by pMLKL oligomerization. Overexpression of W96A-GLTP, an ablated GSL binding site mutant, failed to arrest the cell cycle or induce necroptosis. Sphingolipid assessment (ceramide, monohexosylceramide, sphingomyelin, ceramide-1-phosphate, sphingosine, and sphingosine-1-phosphate) of HT-29 cells overexpressing GLTP revealed large decreases (>5-fold) in sphingosine-1-phosphate with minimal change in 16:0-ceramide, tipping the 'sphingolipid rheostat' (S1P/16:0-Cer ratio) towards cell death. Depletion of RIPK-3 or MLKL abrogated necroptosis induced by GLTP overexpression. Our findings establish GLTP upregulation as a previously unknown suppressor of human colon carcinoma HT-29 cells via interference with cell cycle progression and induction of necroptosis.
Collapse
Affiliation(s)
| | - Daniel J Stephenson
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University Medical Center, Richmond, VA 23298-0614, USA; Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA; Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; The Moffitt Cancer Center, Tampa, FL 33620, USA
| | | |
Collapse
|
33
|
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: 0.8] [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
|
34
|
Kashyap VK, Wang Q, Setua S, Nagesh PKB, Chauhan N, Kumari S, Chowdhury P, Miller DD, Yallapu MM, Li W, Jaggi M, Hafeez BB, Chauhan SC. Therapeutic efficacy of a novel βIII/βIV-tubulin inhibitor (VERU-111) in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:29. [PMID: 30674344 PMCID: PMC6343279 DOI: 10.1186/s13046-018-1009-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
Abstract
Background The management of pancreatic cancer (PanCa) is exceptionally difficult due to poor response to available therapeutic modalities. Tubulins play a major role in cell dynamics, thus are important molecular targets for cancer therapy. Among various tubulins, βIII and βIV-tubulin isoforms have been primarily implicated in PanCa progression, metastasis and chemo-resistance. However, specific inhibitors of these isoforms that have potent anti-cancer activity with low toxicity are not readily available. Methods We determined anti-cancer molecular mechanisms and therapeutic efficacy of a novel small molecule inhibitor (VERU-111) using in vitro (MTS, wound healing, Boyden chamber and real-time xCELLigence assays) and in vivo (xenograft studies) models of PanCa. The effects of VERU-111 treatment on the expression of β-tubulin isoforms, apoptosis, cancer markers and microRNAs were determined by Western blot, immunohistochemistry (IHC), confocal microscopy, qRT-PCR and in situ hybridization (ISH) analyses. Results We have identified a novel small molecule inhibitor (VERU-111), which preferentially represses clinically important, βIII and βIV tubulin isoforms via restoring the expression of miR-200c. As a result, VERU-111 efficiently inhibited tumorigenic and metastatic characteristics of PanCa cells. VERU-111 arrested the cell cycle in the G2/M phase and induced apoptosis in PanCa cell lines via modulation of cell cycle regulatory (Cdc2, Cdc25c, and Cyclin B1) and apoptosis - associated (Bax, Bad, Bcl-2, and Bcl-xl) proteins. VERU-111 treatment also inhibited tumor growth (P < 0.01) in a PanCa xenograft mouse model. Conclusions This study has identified an inhibitor of βIII/βIV tubulins, which appears to have excellent potential as monotherapy or in combination with conventional therapeutic regimens for PanCa treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-1009-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vivek K Kashyap
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Qinghui Wang
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Saini Setua
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Neeraj Chauhan
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Sonam Kumari
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| | - Bilal Bin Hafeez
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
35
|
Guzmán EA. Regulated Cell Death Signaling Pathways and Marine Natural Products That Target Them. Mar Drugs 2019; 17:md17020076. [PMID: 30678065 PMCID: PMC6410226 DOI: 10.3390/md17020076] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Our understanding of cell death used to consist in necrosis, an unregulated form, and apoptosis, regulated cell death. That understanding expanded to acknowledge that apoptosis happens through the intrinsic or extrinsic pathways. Actually, many other regulated cell death processes exist, including necroptosis, a regulated form of necrosis, and autophagy-dependent cell death. We also understand that apoptosis occurs beyond the intrinsic and extrinsic pathways with caspase independent forms of apoptosis existing. Our knowledge of the signaling continues to grow, and with that, so does our ability to target different parts of the pathways with small molecules. Marine natural products co-evolve with their targets, and these unique molecules have complex structures with exquisite biological activities and specificities. This article offers a review of our current understanding of the signaling pathways regulating cell death, and highlights marine natural products that can affect these signaling pathways.
Collapse
Affiliation(s)
- Esther A Guzmán
- Marine Biomedical and Biotechnology Research, Harbor Branch Oceanographic Institute at Florida Atlantic University, 5600 US 1 North, Fort Pierce, FL 34946, USA.
| |
Collapse
|
36
|
Hou W, Fan Q, Su L, Xu H. Synthesis of Oridonin Derivatives via Mizoroki-Heck Reaction and Click Chemistry for Cytotoxic Activity. Anticancer Agents Med Chem 2019; 19:935-947. [PMID: 30657049 DOI: 10.2174/1871520619666190118121439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 01/01/2019] [Accepted: 01/07/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Natural products (NPs) are evolutionarily chosen "privileged structures" that have a profound impact upon the anticancer drug discovery and development progress. However, the search for new drugs based on structure modification of NPs has often been hindered due to the tedious and complicated synthetic pathways. Fortunately, Mizoroki-Heck reaction and copper-catalyzed alkyne-azide cycloaddition (CuAAC) could provide perfect strategies for selective modification on NPs even in the presence of liable functionalities. OBJECTIVE Here, we used oridonin, an ent-kaurane diterpenoid that showed a wide range of biological activities, as a parent molecule for the generation of analogues with anticancer activity. METHODS Derivatives of oridonin were generated based on the structure-activity relationship study of oridonin and synthesized via Mizoroki-Heck reaction and CuAAC. The cytotoxicity of new oridonin derivatives were evaluated on both cancer cells and normal cells. Furthermore, the apoptotic effect and cell cycle arrest effect of the selected potent analogue were evaluated by flow cytometry and western blotting analysis. RESULTS Two series of novel C-14 and C-17 modified derivatives of oridonin were obtained via Heck reaction and copper-catalyzed alkyne-azide cycloaddition (CuAAC), respectively. In vitro antiproliferative activities showed that the introduction of C-14 (2-triazole)acetoxyl- moiety could retain or enhance cytotoxicity, whereas the introduction of C-17 phenyl ring might exert negative effect. Further studies demonstrated that derivative 23 exhibited broad-spectrum antiproliferative activity, effectively overcame drug-resistance and showed weak cytotoxicity on non-cancer cells. Preliminary mechanistic studies indicated that 23 might cause G2/M phase arrest and induce apoptosis in PC-3 cells. CONCLUSION Mizoroki-Heck reaction and CuAAC are perfect strategies for structure modification of complex natural products. The introduction of C-14 (2-triazole)acetoxyl- moiety could retain or enhance the cytotoxicity of oridonin, the introduction of C-17 phenyl group might exert negative effect on its cytotoxicity.
Collapse
Affiliation(s)
- Wei Hou
- College of Pharmaceutical Science, and Institute of Drug Development & Chemical Biology (IDD&CB), Zhejiang University of Technology, Hangzhou, 310014, China
| | - Qiuju Fan
- Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Lin Su
- College of Pharmaceutical Science, and Institute of Drug Development & Chemical Biology (IDD&CB), Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, 201210, China
| |
Collapse
|
37
|
Luo G, Tang Z, Li X, Hou Q, Chen Y, Lao K, Xiang H. 3, 9-di-O-substituted coumestrols incorporating basic amine side chains act as novel apoptosis inducers with improved pharmacological selectivity. Bioorg Chem 2019; 85:140-151. [PMID: 30612080 DOI: 10.1016/j.bioorg.2018.12.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/07/2018] [Accepted: 12/18/2018] [Indexed: 01/11/2023]
Abstract
There is much interest in the use of phytoestrogens such as coumestrol in breast cancer intervention due to their antiestrogenic activity and multiple modes of tumor cell death. However, the clear beneficial effects of naturally occurring estrogen mimetic coumestrol remain controversial due to experimental evidence that it has been shown to stimulate MCF-7 cell proliferation via agonist effect on estrogen receptor at low concentration. Herein, to disconnect the ER interaction and apoptosis-specific mechanism of coumestrol, various 3, 9-di-O-substituted coumestrols (7a-7e) and their furan ring-opened analogs (5a-5e) were synthesized and assessed for antiproliferative properties. Attachment of a dimethylamine-containing side chain to 3-O of coumestrol led to the most promising compound 7e with improved antiproliferative activity (1.7-fold increase) against MCF-7 cells, decreased estrogen activity (>20 times weaker ERα binder) and a novel action to induce apoptosis. Mechanistic studies revealed that 7e is a tubulin polymerization inhibitor, which could arrest cell cycle at G2/M phase and induce apoptosis along with the decrease of mitochondrial membrane potential. In summary, such subtle modifications to the 3, 9-di-hydroxyl groups of coumestrol allow the generation of a novel apoptosis inducer with distinct pharmacological properties, providing an excellent starting point to future development of novel tumor-vascular disrupting agents targeting tubulin.
Collapse
Affiliation(s)
- Guoshun Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhengpu Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xinyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiangqiang Hou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yu Chen
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Smart Drug Delivery, Ministry of Education School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai 201203, China
| | - Kejing Lao
- Shaanxi Key Laboratory of Brain Disorders and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Hua Xiang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
38
|
Design and Synthesis of C-19 Isosteviol Derivatives as Potent and Highly Selective Antiproliferative Agents. Molecules 2018; 24:molecules24010121. [PMID: 30598028 PMCID: PMC6337650 DOI: 10.3390/molecules24010121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 12/24/2018] [Accepted: 12/24/2018] [Indexed: 12/29/2022] Open
Abstract
Six series of novel isosteviol derivatives; modified in the C-19 position; were synthesized; and their antiproliferative activity was evaluated against three human cancer cell lines (HCT-116; BEL-7402; HepG2) and the human L02 normal cell line in vitro. Most of the derivatives tested here exhibited improved antiproliferative activity with high selectivity when compared with the parent compound isosteviol and the positive control drug 5-fluorouracil. Among these derivatives; compound 5d exhibited the most potent antiproliferative activity and commendable selectivity between cancer and normal cells. In addition; compound 5d inhibited the colony formation of HCT-116 cells in a concentration-dependent manner. Further studies revealed that compound 5d arrested the HCT-116 cell cycle in the S phase; and western blot analysis demonstrated the mechanism may be correlated with a change in the expression of cyclin A; cyclin B1; and cyclin E1. Furthermore; the results of a docking study that involved placing compound 5d into the CDK2/cyclin A binding site revealed that its mode of action was possibly as a CDK2/cyclin A inhibitor.
Collapse
|
39
|
Li W, Xu F, Shuai W, Sun H, Yao H, Ma C, Xu S, Yao H, Zhu Z, Yang DH, Chen ZS, Xu J. Discovery of Novel Quinoline–Chalcone Derivatives as Potent Antitumor Agents with Microtubule Polymerization Inhibitory Activity. J Med Chem 2018; 62:993-1013. [DOI: 10.1021/acs.jmedchem.8b01755] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Wenlong Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Feijie Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Wen Shuai
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Honghao Sun
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Cong Ma
- State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Hequan Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| |
Collapse
|
40
|
Xin YB, Li JJ, Zhang HJ, Ma J, Liu X, Gong GH, Tian YS. Synthesis and characterisation of (Z)-styrylbenzene derivatives as potential selective anticancer agents. J Enzyme Inhib Med Chem 2018; 33:1554-1564. [PMID: 30244610 PMCID: PMC6161602 DOI: 10.1080/14756366.2018.1513925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/31/2018] [Accepted: 08/16/2018] [Indexed: 01/06/2023] Open
Abstract
To identify anticancer agents with high potency and low toxicity, a series of (Z)-styrylbenzene derivatives were synthesised and evaluated for anticancer activities using a panel of nine cancer cell lines and two noncancerous cell lines. Most derivatives exhibited significant anti-proliferative activities against five cancer cell lines, including MGC-803 and BEL-7402. (Z)-3-(p-Tolyl)-2-(3,4,5-trimethoxyphenyl)acrylonitrile (6h) showed a strong inhibitory effect on MGC-803 cells (IC50 < 0.01 µM) and exhibited stronger anti-proliferative activity than taxol (IC50 < 0.06 ± 0.01 µM). The IC50 value of 6h in L-02 cells was 10,000-fold higher than in MGC-803 cells. Compound 6h inhibited proliferation of BEL-7402 cells by arresting at the G2/M phase through up-regulation of cyclin B1 expression, down-regulation of cyclin A and D1 expression, and induction of apoptosis. In addition, 6h inhibited the migration of BEL-7402 cells and the formation of cell colonies.
Collapse
Affiliation(s)
- Ya-Bing Xin
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Jia-Jun Li
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Hong-Jian Zhang
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Jun Ma
- Jiangsu Hansoh Pharmaceutical Group Co., Ltd., Lianyungang, P.R. China
| | - Xin Liu
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Guo-Hua Gong
- First Clinical Medical College of Inner Mongolia University for Nationalities, Tongliao, P.R. China
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Inner Mongolia University for Nationalities, Tongliao, P.R. China
| | - Yu-Shun Tian
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| |
Collapse
|
41
|
Hou W, Zhang G, Luo Z, Su L, Xu H. Click chemistry‐based synthesis and cytotoxic activity evaluation of 4α‐triazole acetate podophyllotoxin derivatives. Chem Biol Drug Des 2018; 93:473-483. [DOI: 10.1111/cbdd.13436] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 09/26/2018] [Accepted: 10/28/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Wei Hou
- College of Pharmaceutical ScienceInstitute of Drug Development & Chemical Biology (IDD & CB)Zhejiang University of Technology Hangzhou China
| | - Guanjun Zhang
- College of Chemical Engineering and Materials ScienceTianjin University of Science & Technology Tianjin China
| | - Zhi Luo
- Shanghai Evergene Biotech Co., Ltd. Shanghai China
| | - Lin Su
- College of Pharmaceutical ScienceInstitute of Drug Development & Chemical Biology (IDD & CB)Zhejiang University of Technology Hangzhou China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech University Shanghai China
| |
Collapse
|
42
|
Vicente-Blázquez A, González M, Álvarez R, Del Mazo S, Medarde M, Peláez R. Antitubulin sulfonamides: The successful combination of an established drug class and a multifaceted target. Med Res Rev 2018; 39:775-830. [PMID: 30362234 DOI: 10.1002/med.21541] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/02/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022]
Abstract
Tubulin, the microtubules and their dynamic behavior are amongst the most successful antitumor, antifungal, antiparasitic, and herbicidal drug targets. Sulfonamides are exemplary drugs with applications in the clinic, in veterinary and in the agrochemical industry. This review summarizes the actual state and recent progress of both fields looking from the double point of view of the target and its drugs, with special focus onto the structural aspects. The article starts with a brief description of tubulin structure and its dynamic assembly and disassembly into microtubules and other polymers. Posttranslational modifications and the many cellular means of regulating and modulating tubulin's biology are briefly presented in the tubulin code. Next, the structurally characterized drug binding sites, their occupying drugs and the effects they induce are described, emphasizing on the structural requirements for high potency, selectivity, and low toxicity. The second part starts with a summary of the favorable and highly tunable combination of physical-chemical and biological properties that render sulfonamides a prototypical example of privileged scaffolds with representatives in many therapeutic areas. A complete description of tubulin-binding sulfonamides is provided, covering the different species and drug sites. Some of the antimitotic sulfonamides have met with very successful applications and others less so, thus illustrating the advances, limitations, and future perspectives of the field. All of them combine in a mechanism of action and a clinical outcome that conform efficient drugs.
Collapse
Affiliation(s)
- Alba Vicente-Blázquez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Myriam González
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Sara Del Mazo
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Manuel Medarde
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Facultad de Farmacia, Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| |
Collapse
|
43
|
Evaluation of 4-phenylamino-substituted naphthalene-1,2-diones as tubulin polymerization inhibitors. Bioorg Med Chem Lett 2018; 28:3057-3063. [DOI: 10.1016/j.bmcl.2018.07.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 11/24/2022]
|
44
|
Hip2 ubiquitin-conjugating enzyme has a role in UV-induced G1/S arrest and re-entry. Genes Genomics 2018; 41:159-166. [PMID: 30264212 DOI: 10.1007/s13258-018-0747-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022]
Abstract
Regulation of cell cycle arrest and re-entry triggered by DNA damage is vital for cell division and growth and is also involved in cell survival. UV radiation can generate lesions in the DNA, which results in cell cycle arrest and the induction of the DNA repair process. However, the mechanism of promoting cell cycle progression following DNA repair is elusive. The primary aim of this study is to investigate whether Hip2 ubiquitin-conjugating enzyme has a role in UV-induced G1/S arrest and re-entry. The phase of HEK293 cells was synchronized at the G1/S border using thymidine. The synchronously proliferating cells were exposed to UV radiation to cause DNA damage. We investigated the expression of p53, Hip2, p21, cyclin D and E proteins that are involved in the cell cycle progression. Finally, we examined changes in the phosphorylation of Hip2 after UV radiation treatment using the pIMAGO™ assay. When cells were exposed to UV radiation, expression of p53 was elevated, and the cell cycle was arrested at the G1/S boundary. In response to the increased p53 level, Hip2 became phosphorylated and activated through the inhibition of its degradation. The phosphorylated Hip2 inhibited p53, thereby suppressing the expression of p21, a downstream signal, and sequentially stimulating cyclin D and cyclin E to induce re-entry to the cell cycle. Our studies demonstrate that Hip2 works as a regulator in UV-induced cell cycle arrest and re-entry.
Collapse
|
45
|
Allan LA, Skowyra A, Rogers KI, Zeller D, Clarke PR. Atypical APC/C-dependent degradation of Mcl-1 provides an apoptotic timer during mitotic arrest. EMBO J 2018; 37:e96831. [PMID: 29987118 PMCID: PMC6120658 DOI: 10.15252/embj.201796831] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/03/2018] [Accepted: 05/17/2018] [Indexed: 12/28/2022] Open
Abstract
The initiation of apoptosis in response to the disruption of mitosis provides surveillance against chromosome instability. Here, we show that proteolytic destruction of the key regulator Mcl-1 during an extended mitosis requires the anaphase-promoting complex or cyclosome (APC/C) and is independent of another ubiquitin E3 ligase, SCFFbw7 Using live-cell imaging, we show that the loss of Mcl-1 during mitosis is dependent on a D box motif found in other APC/C substrates, while an isoleucine-arginine (IR) C-terminal tail regulates the manner in which Mcl-1 engages with the APC/C, converting Mcl-1 from a Cdc20-dependent and checkpoint-controlled substrate to one that is degraded independently of checkpoint strength. This mechanism ensures a relatively slow but steady rate of Mcl-1 degradation during mitosis and avoids its catastrophic destruction when the mitotic checkpoint is satisfied, providing an apoptotic timer that can distinguish a prolonged mitotic delay from normal mitosis. Importantly, we also show that inhibition of Cdc20 promotes mitotic cell death more effectively than loss of APC/C activity through differential effects on Mcl-1 degradation, providing an improved strategy to kill cancer cells.
Collapse
Affiliation(s)
- Lindsey A Allan
- Division of Cancer Research, Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Agnieszka Skowyra
- Division of Cancer Research, Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Katie I Rogers
- Division of Cancer Research, Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Désirée Zeller
- Division of Cancer Research, Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Paul R Clarke
- Division of Cancer Research, Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- The University of Queensland Diamantina Institute Faculty of Medicine Translational Research Institute, Woolloongabba, Qld, Australia
| |
Collapse
|
46
|
Discovery of novel vinyl sulfone derivatives as anti-tumor agents with microtubule polymerization inhibitory and vascular disrupting activities. Eur J Med Chem 2018; 157:1068-1080. [DOI: 10.1016/j.ejmech.2018.08.074] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/07/2018] [Accepted: 08/26/2018] [Indexed: 11/18/2022]
|
47
|
Lynce F, Shajahan-Haq AN, Swain SM. CDK4/6 inhibitors in breast cancer therapy: Current practice and future opportunities. Pharmacol Ther 2018; 191:65-73. [PMID: 29933034 DOI: 10.1016/j.pharmthera.2018.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dysregulation of the cyclin dependent kinase pathway in luminal breast cancer creates a new therapeutic opportunity for estrogen receptor positive breast cancer. Initial pan-CDK inhibitors were associated with extensive toxicities but in recent years, the development of potent specific CDK inhibitors with favorable tolerability has driven renewed interests in this class of targeted therapies. Palbociclib, ribociclib and abemaciclib are specific CDK4/6 inhibitors that have been approved by the U.S. Food and Drug Administration for use in combination with endocrine therapy for women with advanced hormone receptor positive breast cancer. These three anticancer therapeutics were approved based on progression free survival benefit seen on phase III trials with the most common grade 3 treatment-related side effects being neutropenia, fatigue, nausea and diarrhea. Except for estrogen receptor positivity, no biomarkers predictive of response to CDK4/6 inhibitors have been identified to date. Based on mechanistic insights here described, CDK4/6 inhibitors are currently being explored in combination with other agents, including targeted therapies, immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Filipa Lynce
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Ayesha N Shajahan-Haq
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Sandra M Swain
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
48
|
Crawford N, Salvucci M, Hellwig CT, Lincoln FA, Mooney RE, O'Connor CL, Prehn JH, Longley DB, Rehm M. Simulating and predicting cellular and in vivo responses of colon cancer to combined treatment with chemotherapy and IAP antagonist Birinapant/TL32711. Cell Death Differ 2018; 25:1952-1966. [PMID: 29500433 DOI: 10.1038/s41418-018-0082-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/30/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022] Open
Abstract
Apoptosis resistance contributes to treatment failure in colorectal cancer (CRC). New treatments that reinstate apoptosis competency have potential to improve patient outcome but require predictive biomarkers to target them to responsive patient populations. Inhibitor of apoptosis proteins (IAPs) suppress apoptosis, contributing to drug resistance; IAP antagonists such as TL32711 have therefore been developed. We developed a systems biology approach for predicting response of CRC cells to chemotherapy and TL32711 combinations in vitro and in vivo. CRC cells responded poorly to TL32711 monotherapy in vitro; however, co-treatment with 5-fluorouracil (5-FU) and oxaliplatin enhanced TL32711-induced apoptosis. Notably, cells from genetically identical populations responded highly heterogeneously, with caspases being activated both upstream and downstream of mitochondrial outer membrane permeabilisation (MOMP). These data, combined with quantities of key apoptosis regulators were sufficient to replicate in vitro cell death profiles by mathematical modelling. In vivo, apoptosis protein expression was significantly altered, and mathematical modelling for these conditions predicted higher apoptosis resistance that could nevertheless be overcome by combination of chemotherapy and TL32711. Subsequent experimental observations agreed with these predictions, and the observed effects on tumour growth inhibition correlated robustly with apoptosis competency. We therefore obtained insights into intracellular signal transduction kinetics and their population-based heterogeneities for chemotherapy/TL32711 combinations and provide proof-of-concept that mathematical modelling of apoptosis competency can simulate and predict responsiveness in vivo. Being able to predict response to IAP antagonist-based treatments on the background of cell-to-cell heterogeneities in the future might assist in improving treatment stratification approaches for these emerging apoptosis-targeting agents.
Collapse
Affiliation(s)
- Nyree Crawford
- Cell Death & Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Manuela Salvucci
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Christian T Hellwig
- Institute of Cell Biology and Immunology, University of Stuttgart, D-70569, Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, D-70569, Stuttgart, Germany
| | - Frank A Lincoln
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ruth E Mooney
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Carla L O'Connor
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jochen Hm Prehn
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Daniel B Longley
- Cell Death & Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK.
| | - Markus Rehm
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland. .,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland. .,Institute of Cell Biology and Immunology, University of Stuttgart, D-70569, Stuttgart, Germany. .,Stuttgart Research Center Systems Biology, University of Stuttgart, D-70569, Stuttgart, Germany.
| |
Collapse
|
49
|
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: 2.2] [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
|
50
|
An B, Zhang S, Hu J, Pan T, Huang L, Tang JCO, Li X, Chan ASC. The design, synthesis and evaluation of selenium-containing 4-anilinoquinazoline hybrids as anticancer agents and a study of their mechanism. Org Biomol Chem 2018; 16:4701-4714. [DOI: 10.1039/c8ob00875b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We report the design, synthesis and evaluation of 2-chloro-N-methyl-N-(4-selenocyanatophenyl)quinazolin-4-amine and its analogues as anticancer agents and the mechanism study.
Collapse
Affiliation(s)
- Baijiao An
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Shun Zhang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Jinhui Hu
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Tingting Pan
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Ling Huang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Johnny Cheuk-on Tang
- State Key Laboratory of Chirosciences
- Department of Applied Biology and Chemical Technology
- The Hong Kong Polytechnic University
- P.R. China
| | - Xingshu Li
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Albert S. C. Chan
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| |
Collapse
|