1
|
Müller J, Boubaker G, Müller N, Uldry AC, Braga-Lagache S, Heller M, Hemphill A. Investigating Antiprotozoal Chemotherapies with Novel Proteomic Tools-Chances and Limitations: A Critical Review. Int J Mol Sci 2024; 25:6903. [PMID: 39000012 PMCID: PMC11241152 DOI: 10.3390/ijms25136903] [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: 05/31/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Identification of drug targets and biochemical investigations on mechanisms of action are major issues in modern drug development. The present article is a critical review of the classical "one drug"-"one target" paradigm. In fact, novel methods for target deconvolution and for investigation of resistant strains based on protein mass spectrometry have shown that multiple gene products and adaptation mechanisms are involved in the responses of pathogens to xenobiotics rather than one single gene or gene product. Resistance to drugs may be linked to differential expression of other proteins than those interacting with the drug in protein binding studies and result in complex cell physiological adaptation. Consequently, the unraveling of mechanisms of action needs approaches beyond proteomics. This review is focused on protozoan pathogens. The conclusions can, however, be extended to chemotherapies against other pathogens or cancer.
Collapse
Affiliation(s)
- Joachim Müller
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Ghalia Boubaker
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Norbert Müller
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Anne-Christine Uldry
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Sophie Braga-Lagache
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| |
Collapse
|
2
|
Zhang W, Dai J, Hou G, Liu H, Zheng S, Wang X, Lin Q, Zhang Y, Lu M, Gong Y, Xiang Z, Yu Y, Hu Y. SMURF2 predisposes cancer cell toward ferroptosis in GPX4-independent manners by promoting GSTP1 degradation. Mol Cell 2023; 83:4352-4369.e8. [PMID: 38016474 DOI: 10.1016/j.molcel.2023.10.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 09/22/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
Ferroptosis is a non-apoptotic form of regulated cell death. Glutathione (GSH) peroxidase 4 (GPX4) and GSH-independent ferroptosis suppressor protein 1 (FSP1) have been identified as major defenses. Here, we uncover a protective mechanism mediated by GSH S-transferase P1 (GSTP1) by monitoring proteinomic dynamics during ferroptosis. Dramatic downregulation of GSTP1 is caused by SMURF2-mediated GSTP1 ubiquitination and degradation at early stages of ferroptosis. Intriguingly, GSTP1 acts in GPX4- and FSP1-independent manners by catalyzing GSH conjugation of 4-hydroxynonenal and detoxifying lipid hydroperoxides via selenium-independent GSH peroxidase activity. Genetic modulation of the SMURF2/GSTP1 axis or the pharmacological inhibition of GSTP1's catalytic activity sensitized tumor responses to Food and Drug Administration (FDA)-approved ferroptosis-inducing drugs both in vitro and in vivo. GSTP1 expression also confers resistance to immune checkpoint inhibitors by blunting ferroptosis. Collectively, these findings demonstrate a GPX4/FSP1-independent cellular defense mechanism against ferroptosis and suggest that targeting SMURF2/GSTP1 to sensitize cancer cells to ferroptosis has potential as an anticancer therapy.
Collapse
Affiliation(s)
- Wenxin Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou 450000, China
| | - Junren Dai
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | | | - Hao Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Shanliang Zheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Qingyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yi Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Minqiao Lu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yafan Gong
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Zhiyuan Xiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yan Yu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou 450000, China.
| |
Collapse
|
3
|
Imbaby S, Elkholy SE, Faisal S, Abdelmaogood AKK, Mehana AE, Mansour BSA, Abd El-Moneam SM, Elaidy SM. The GSTP1/MAPKs/BIM/SMAC modulatory actions of nitazoxanide: Bioinformatics and experimental evidence in subcutaneous solid Ehrlich carcinoma-inoculated mice. Life Sci 2023; 319:121496. [PMID: 36822315 DOI: 10.1016/j.lfs.2023.121496] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/25/2023]
Abstract
AIMS Ehrlich ascites carcinoma and its subcutaneous inoculated solid tumour form (SEC) are reliable models for chemotherapeutic molecular targets exploration. Novel chemotherapeutic approaches are identified as molecular targets for intrinsic apoptosis, like the modulation of the second mitochondria-derived activator of caspases (SMAC). SMAC is a physiological substrate of mitogen-activated protein kinases (MAPKs). Glutathione-S-transferase P1 (GSTP1) and its close association with MAPKs play an important role in malignant cell proliferation, metastasis, and resistance to chemotherapeutics. Nitazoxanide (NTZ) is an emerging cancer therapy and its targeted GSTP1 evidence remains a knowledge need. MAIN METHODS In the present mice-established SEC, the chemotherapeutic roles of oral NTZ (200 mg/kg/day) and 5-fluorouracil (5-FU; 20 mg/kg/day, intraperitoneally) regimens were evaluated by measuring changes in tumour mass, the tumour MAPKs, cytochrome c, Bcl-2 interacting mediator of cell death (BIM), and SMAC signalling pathway in addition to its molecular downstream; caspases 3 and 9. KEY FINDINGS Computational analysis for these target protein interactions showed direct-ordered interactions. After individual therapy with NTZ and 5-FU regimens, the histological architecture of the extracted tumour discs revealed decreases in viable tumour regions with significant necrosis surrounds. These findings were consistent with gross tumour sizes. Each separate regimen lowered the remarkable GSTP1 and elevated the low MAPKs expressions, cytochrome c, BIM, SMAC, and caspases 3, and 9 in EST tissues. SIGNIFICANCE The chemotherapeutic activity of NTZ in SEC was proven. Additionally, NTZ possesses a SMAC modulatory activity that, following thorough research, should be taken into consideration as a chemotherapeutic approach in solid tumours.
Collapse
Affiliation(s)
- Samar Imbaby
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt.
| | - Shereen E Elkholy
- Department of Clinical Pharmacology, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Salwa Faisal
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Asmaa K K Abdelmaogood
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Amir E Mehana
- Department of Zoology, Faculty of Science, Suez Canal University, 41522 Ismailia, Egypt
| | - Basma S A Mansour
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Samar M Abd El-Moneam
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Samah M Elaidy
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt.
| |
Collapse
|
4
|
Lü Z, Li X, Li K, Ripani P, Shi X, Xu F, Wang M, Zhang L, Brunner T, Xu P, Niu Y. Nitazoxanide and related thiazolides induce cell death in cancer cells by targeting the 20S proteasome with novel binding modes. Biochem Pharmacol 2022; 197:114913. [PMID: 35032461 DOI: 10.1016/j.bcp.2022.114913] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/08/2021] [Accepted: 01/05/2022] [Indexed: 11/02/2022]
Abstract
Nitazoxanide and related thiazolides are a novel class of anti-infectious agents against protozoan parasites, bacteria and viruses. In recent years, it is demonstrated thatthiazolides also can induced cell cycle arrest and apoptotic cell death in cancer cells. Due to their fast proliferating nature, cancer cells highly depend on the proteasome system to remove aberrant proteins. Many of these aberrant proteins are regulators of cell cycle progression and apoptosis, such as the cyclins, BCL2 family members and nuclear factor of κB (NF-κB). Here, we demonstrate at both molecular and cellular levels that the 20S proteasome is a direct target of NTZ and other thiazolides. By concurrently inhibiting the multiple catalytic subunits of 20S proteasome, NTZ promotes cell cycle arrest and triggers cell death in colon cancer cells, either directly or a sensitizer to other anti-tumor agents, especially doxorubicin. We further show that the binding mode of NTZ in the β5 subunit of the 20S proteasome is different from that of bortezomib and other existing proteasome inhibitors. These findings provide new insights in the design of novel small molecular proteasome inhibitors as anti-tumor agents suitable for solid tumor treatment in an oral dosing form.
Collapse
Affiliation(s)
- Zirui Lü
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China
| | - Xiaona Li
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China
| | - Kebin Li
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China
| | - Paola Ripani
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstr. 10, 78457 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457 Konstanz, Germany
| | - Xiaomeng Shi
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China
| | - Fengrong Xu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China
| | - Mopei Wang
- Department of Tumor Chemotherapy and Radiation Sickness, Peking University Third Hospital, Beijing, 100191, China
| | - Liangren Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstr. 10, 78457 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstr. 10, 78457 Konstanz, Germany
| | - Ping Xu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China.
| | - Yan Niu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University Health Science Center, Xueyuan Road 38, Beijing, 100191, China.
| |
Collapse
|
5
|
Ahmed T, Rahman SMA, Asaduzzaman M, Islam ABMMK, Chowdhury AKA. Synthesis, in vitro bioassays, and computational study of heteroaryl nitazoxanide analogs. Pharmacol Res Perspect 2021; 9:e00800. [PMID: 34086411 PMCID: PMC8177060 DOI: 10.1002/prp2.800] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/03/2021] [Indexed: 12/20/2022] Open
Abstract
Antiprotozoal drug nitazoxanide (NTZ) has shown diverse pharmacological properties and has appeared in several clinical trials. Herein we present the synthesis, characterization, in vitro biological investigation, and in silico study of four hetero aryl amide analogs of NTZ. Among the synthesized molecules, compound 2 and compound 4 exhibited promising antibacterial activity against Escherichia coli (E. coli), superior to that displayed by the parent drug nitazoxanide as revealed from the in vitro antibacterial assay. Compound 2 displayed zone of inhibition of 20 mm, twice as large as the parent drug NTZ (10 mm) in their least concentration (12.5 µg/ml). Compound 1 also showed antibacterial effect similar to that of nitazoxanide. The analogs were also tested for in vitro cytotoxic activity by employing cell counting kit-8 (CCK-8) assay technique in HeLa cell line, and compound 2 was identified as a potential anticancer agent having IC50 value of 172 µg which proves it to be more potent than nitazoxanide (IC50 = 428 µg). Furthermore, the compounds were subjected to molecular docking study against various bacterial and cancer signaling proteins. The in vitro test results corroborated with the in silico docking study as compound 2 and compound 4 had comparatively stronger binding affinity against the proteins and showed a higher docking score than nitazoxanide toward human mitogen-activated protein kinase (MAPK9) and fatty acid biosynthesis enzyme (FabH) of E. coli. Moreover, the docking study demonstrated dihydrofolate reductase (DHFR) and thymidylate synthase (TS) as probable new targets for nitazoxanide and its synthetic analogs. Overall, the study suggests that nitazoxanide and its analogs can be a potential lead compound in the drug development.
Collapse
Affiliation(s)
- Tasmia Ahmed
- Department of Clinical Pharmacy and PharmacologyFaculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - S. M. Abdur Rahman
- Department of Clinical Pharmacy and PharmacologyFaculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - Muhammad Asaduzzaman
- Department of Clinical Pharmacy and PharmacologyFaculty of PharmacyUniversity of DhakaDhakaBangladesh
| | | | - A. K. Azad Chowdhury
- Department of Clinical Pharmacy and PharmacologyFaculty of PharmacyUniversity of DhakaDhakaBangladesh
| |
Collapse
|
6
|
Michalek S, Brunner T. Nuclear-mitochondrial crosstalk: On the role of the nuclear receptor liver receptor homolog-1 (NR5A2) in the regulation of mitochondrial metabolism, cell survival, and cancer. IUBMB Life 2020; 73:592-610. [PMID: 32931651 DOI: 10.1002/iub.2386] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
Liver receptor homolog-1 (LRH-1, NR5A2) is an orphan nuclear receptor with widespread activities in the regulation of development, stemness, metabolism, steroidogenesis, and proliferation. Many of the LRH-1-regulated processes target the mitochondria and associated activities. While under physiological conditions, a balanced LRH-1 expression and regulation contribute to the maintenance of a physiological equilibrium, deregulation of LRH-1 has been associated with inflammation and cancer. In this review, we discuss the role and mechanism(s) of how LRH-1 regulates metabolic processes, cell survival, and cancer in a nuclear-mitochondrial crosstalk, and evaluate its potential as a pharmacological target.
Collapse
Affiliation(s)
- Svenja Michalek
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
7
|
Qiu K, Zheng Z, Huang Y. Long intergenic noncoding RNA 00844 promotes apoptosis and represses proliferation of prostate cancer cells through upregulating GSTP1 by recruiting EBF1. J Cell Physiol 2020; 235:8472-8485. [PMID: 32329523 DOI: 10.1002/jcp.29690] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/03/2020] [Accepted: 03/24/2020] [Indexed: 12/24/2022]
Abstract
Accumulating evidence have suggested the function of long noncoding RNAs as crucial players in the pathogenesis of prostate cancer (PC), a urologic tumor in male with poor prognosis. This study was designed to explore the functions of long intergenic noncoding RNA 00844 (LINC00844) in PC progression. The expression of LINC00844 and glutathione S-transferase P1-1 (GSTP1) was detected by reverse transcription quantitative polymerase chain reaction, followed by the identification of the relationship among LINC00844, GSTP1, and early B cell factor 1 (EBF1) by dual luciferase reporter gene assay, RNA immunoprecipitation assay, electrophoretic mobility shift assay, and chromatin immunoprecipitation assay. Using loss- and gain-of-function assays, the effects of LINC00844, GSTP1, and EBF1 on the biological characteristics of PC cells were assessed by cell counting kit-8 assay, 5-ethynyl-2'-deoxyuridine assay, and flow cytometry. Lastly, the results from in vitro experiments were verified in vivo by establishing a xenograft tumor model in nude mice. LINC00844 and GSTP1 both displayed low expression in PC tissues and cells. LINC00844 positively regulated the expression of GSTP1 via recruiting EBF1. Overexpression of LINC00844 reduced proliferation and elevated apoptosis of PC cells through recruiting EBF1, which subsequently upregulated GSTP1. In vivo experiments confirmed that LINC00844 or GSTP1 upregulation attenuated tumor growth. LINC00844 elevated GSTP1 expression by recruiting EBF1 to the promoter region of GSTP1, thereby suppressing PC progression. Hence, LINC00844 is a novel therapeutic target for the development of new treatment protocols for PC.
Collapse
Affiliation(s)
- Kaiyan Qiu
- Department of Urology, Quanzhou First Hospital Affiliated Fujian Medical University, Quanzhou, P.R. China
| | - Zhixiong Zheng
- Department of Urology, Quanzhou First Hospital Affiliated Fujian Medical University, Quanzhou, P.R. China
| | - Yingfu Huang
- Department of Urology, Quanzhou First Hospital Affiliated Fujian Medical University, Quanzhou, P.R. China
| |
Collapse
|
8
|
Ripani P, Delp J, Bode K, Delgado ME, Dietrich L, Betzler VM, Yan N, von Scheven G, Mayer TU, Leist M, Brunner T. Thiazolides promote G1 cell cycle arrest in colorectal cancer cells by targeting the mitochondrial respiratory chain. Oncogene 2019; 39:2345-2357. [PMID: 31844249 DOI: 10.1038/s41388-019-1142-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 01/10/2023]
Abstract
Systemic toxicity and tumor cell resistance still limit the efficacy of chemotherapy in colorectal cancer. Therefore, alternative treatments are desperately needed. The thiazolide Nitazoxanide (NTZ) is an FDA-approved drug for the treatment of parasite-mediated infectious diarrhea with a favorable safety profile. Interestingly, NTZ and the thiazolide RM4819-its bromo-derivative lacking antibiotic activity-are also promising candidates for cancer treatment. Yet the exact anticancer mechanism(s) of these compounds still remains unclear. In this study, we systematically investigated RM4819 and NTZ in 2D and 3D colorectal cancer culture systems. Both compounds strongly inhibited proliferation of colon carcinoma cell lines by promoting G1 phase cell cycle arrest. Thiazolide-induced cell cycle arrest was independent of the p53/p21 axis, but was mediated by inhibition of protein translation via the mTOR/c-Myc/p27 pathway, likely caused by inhibition of mitochondrial respiration. While both thiazolides demonstrated mitochondrial uncoupling activity, only RM4819 inhibited the mitochondrial respiratory chain complex III. Interestingly, thiazolides also potently inhibited the growth of murine colonic tumoroids in a comparable manner with cisplatin, while in contrast to cisplatin thiazolides did not affect the growth of primary intestinal organoids. Thus, thiazolides appear to have a tumor-selective antiproliferative activity, which offers new perspectives in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- P Ripani
- Department of Biology, Biochemical Pharmacology, University of Konstanz, Konstanz, Germany.,Konstanz Research School Chemical Biology KORS-CB, University of Konstanz, Konstanz, Germany
| | - J Delp
- Konstanz Research School Chemical Biology KORS-CB, University of Konstanz, Konstanz, Germany.,Chair for In Vitro Toxicology and Biomedicine, inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - K Bode
- Department of Biology, Biochemical Pharmacology, University of Konstanz, Konstanz, Germany
| | - M E Delgado
- Department of Biology, Biochemical Pharmacology, University of Konstanz, Konstanz, Germany
| | - L Dietrich
- Department of Biology, Biochemical Pharmacology, University of Konstanz, Konstanz, Germany
| | - V M Betzler
- Department of Biology, Biochemical Pharmacology, University of Konstanz, Konstanz, Germany.,Biotechnology Institute Thurgau, University of Konstanz, Konstanz, Germany
| | - N Yan
- Department of Medicinal Chemistry, Peking University Health Science Centre, Beijing, China
| | - G von Scheven
- Department of Biology, Molecular Toxicology Group, University of Konstanz, Konstanz, Germany
| | - T U Mayer
- Konstanz Research School Chemical Biology KORS-CB, University of Konstanz, Konstanz, Germany.,Department of Biology, Molecular Genetics, University of Konstanz, Konstanz, Germany
| | - M Leist
- Konstanz Research School Chemical Biology KORS-CB, University of Konstanz, Konstanz, Germany.,Chair for In Vitro Toxicology and Biomedicine, inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - T Brunner
- Department of Biology, Biochemical Pharmacology, University of Konstanz, Konstanz, Germany. .,Konstanz Research School Chemical Biology KORS-CB, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
9
|
Beyerle J, Holowatyj AN, Haffa M, Frei E, Gigic B, Schrotz-King P, Boehm J, Habermann N, Stiborova M, Scherer D, Kölsch T, Skender S, Becker N, Herpel E, Schneider M, Ulrich A, Schirmacher P, Chang-Claude J, Brenner H, Hoffmeister M, Haug U, Owen RW, Ulrich CM. Expression Patterns of Xenobiotic-Metabolizing Enzymes in Tumor and Adjacent Normal Mucosa Tissues among Patients with Colorectal Cancer: The ColoCare Study. Cancer Epidemiol Biomarkers Prev 2019; 29:460-469. [PMID: 31740522 DOI: 10.1158/1055-9965.epi-19-0449] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/15/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Xenobiotic-metabolizing enzymes (XME) play a critical role in the activation and detoxification of several carcinogens. However, the role of XMEs in colorectal carcinogenesis is unclear. METHODS We investigated the expression of XMEs in human colorectal tissues among patients with stage I-IV colorectal cancer (n = 71) from the ColoCare Study. Transcriptomic profiling using paired colorectal tumor and adjacent normal mucosa tissues of XMEs (GSTM1, GSTA1, UGT1A8, UGT1A10, CYP3A4, CYP2C9, GSTP1, and CYP2W1) by RNA microarray was compared using Wilcoxon rank-sum tests. We assessed associations between clinicopathologic, dietary, and lifestyle factors and XME expression with linear regression models. RESULTS GSTM1, GSTA1, UGT1A8, UGT1A10, and CYP3A4 were all statistically significantly downregulated in colorectal tumor relative to normal mucosa tissues (all P ≤ 0.03). Women had significantly higher expression of GSTM1 in normal tissues compared with men (β = 0.37, P = 0.02). By tumor site, CYP2C9 expression was lower in normal mucosa among patients with rectal cancer versus colon cancer cases (β = -0.21, P = 0.0005). Smokers demonstrated higher CYP2C9 expression levels in normal mucosa (β = 0.17, P = 0.02) when compared with nonsmokers. Individuals who used NSAIDs had higher GSTP1 tumor expression compared with non-NSAID users (β = 0.17, P = 0.03). Higher consumption of cooked vegetables (>1×/week) was associated with higher CYP3A4 expression in colorectal tumor tissues (β = 0.14, P = 0.007). CONCLUSIONS XMEs have lower expression in colorectal tumor relative to normal mucosa tissues and may modify colorectal carcinogenesis via associations with clinicopathologic, lifestyle, and dietary factors. IMPACT Better understanding into the role of drug-metabolizing enzymes in colorectal cancer may reveal biological differences that contribute to cancer development, as well as treatment response, leading to clinical implications in colorectal cancer prevention and management.
Collapse
Affiliation(s)
- Jolantha Beyerle
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Andreana N Holowatyj
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah.,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Mariam Haffa
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Eva Frei
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Petra Schrotz-King
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Juergen Boehm
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah.,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Nina Habermann
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Dominique Scherer
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Torsten Kölsch
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Stephanie Skender
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Nikolaus Becker
- NCT Cancer Registry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, University Hospital, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | | | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Haug
- Department of Clinical Epidemiology, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany.,Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany
| | - Robert W Owen
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Cornelia M Ulrich
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah. .,Huntsman Cancer Institute, Salt Lake City, Utah.,Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
10
|
Hearn JM, Hughes GM, Romero-Canelón I, Munro AF, Rubio-Ruiz B, Liu Z, Carragher NO, Sadler PJ. Pharmaco-genomic investigations of organo-iridium anticancer complexes reveal novel mechanism of action. Metallomics 2019; 10:93-107. [PMID: 29131211 DOI: 10.1039/c7mt00242d] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Resistance to platinum drugs (used in >50% of cancer chemotherapies) is a clinical problem. Other precious metal complexes with distinct mechanisms of action might overcome this. Half-sandwich organometallic complexes containing arene or cyclopentadienyl (Cp) ligands show promise. We screened two iridium(iii) complexes [Ir(CpXbiph)(ppy)Cl] (ZL49, 1, ppy = phenylpyridine) and [Ir(CpXph)(azpyNMe2)Cl]PF6 (ZL109, 2, azpyNMe2 = N,N-dimethylphenylazopyridine) in 916 cancer cell lines from 28 tissue types. On average, complex 2 was 78× more potent than 1, 36× more active than cisplatin (CDDP), and strongly active (nanomolar) in patient-derived ovarian cancer cell lines. RNA sequencing of A2780 ovarian cells revealed upregulation of antioxidant responses (NRF2, AP-1) consistent with observed induction of reactive oxygen species (ROS). Protein microarrays, high content imaging and cell cycle analysis showed S/G2 arrest, and late-stage DNA damage response without p53 requirement. The triple-negative breast cancer cell line OCUB-M was highly sensitive to 2 as were cell lines with KIT mutations. Complex 2 exhibits a markedly different pattern of antiproliferative activity compared to the 253 drugs in the Sanger Cancer Genome database, but is most similar to osmium(ii) arene complexes which share the same azopyridine ligand. Redox modulation and DNA damage can provide a multi-targeting strategy, allowing compounds such as 2 to overcome cellular resistance to platinum anticancer drugs.
Collapse
|
11
|
Vale N, Correia-Branco A, Patrício B, Duarte D, Martel F. In vitro studies on the inhibition of colon cancer by amino acid derivatives of bromothiazole. Bioorg Med Chem Lett 2017; 27:3507-3510. [DOI: 10.1016/j.bmcl.2017.05.073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/20/2017] [Accepted: 05/22/2017] [Indexed: 01/18/2023]
|
12
|
Tjioe KC, Tostes Oliveira D, Gavard J. Luteolin Impacts on the DNA Damage Pathway in Oral Squamous Cell Carcinoma. Nutr Cancer 2016; 68:838-47. [DOI: 10.1080/01635581.2016.1180411] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Thiazolides promote apoptosis in colorectal tumor cells via MAP kinase-induced Bim and Puma activation. Cell Death Dis 2015; 6:e1778. [PMID: 26043078 PMCID: PMC4669824 DOI: 10.1038/cddis.2015.137] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/30/2015] [Accepted: 04/01/2015] [Indexed: 12/21/2022]
Abstract
While many anticancer therapies aim to target the death of tumor cells, sophisticated resistance mechanisms in the tumor cells prevent cell death induction. In particular enzymes of the glutathion-S-transferase (GST) family represent a well-known detoxification mechanism, which limit the effect of chemotherapeutic drugs in tumor cells. Specifically, GST of the class P1 (GSTP1-1) is overexpressed in colorectal tumor cells and renders them resistant to various drugs. Thus, GSTP1-1 has become an important therapeutic target. We have recently shown that thiazolides, a novel class of anti-infectious drugs, induce apoptosis in colorectal tumor cells in a GSTP1-1-dependent manner, thereby bypassing this GSTP1-1-mediated drug resistance. In this study we investigated in detail the underlying mechanism of thiazolide-induced apoptosis induction in colorectal tumor cells. Thiazolides induce the activation of p38 and Jun kinase, which is required for thiazolide-induced cell death. Activation of these MAP kinases results in increased expression of the pro-apoptotic Bcl-2 homologs Bim and Puma, which inducibly bind and sequester Mcl-1 and Bcl-xL leading to the induction of the mitochondrial apoptosis pathway. Of interest, while an increase in intracellular glutathione levels resulted in increased resistance to cisplatin, it sensitized colorectal tumor cells to thiazolide-induced apoptosis by promoting increased Jun kinase activation and Bim induction. Thus, thiazolides may represent an interesting novel class of anti-tumor agents by specifically targeting tumor resistance mechanisms, such as GSTP1-1.
Collapse
|
14
|
Michels J, Obrist F, Castedo M, Vitale I, Kroemer G. PARP and other prospective targets for poisoning cancer cell metabolism. Biochem Pharmacol 2014; 92:164-71. [PMID: 25199458 DOI: 10.1016/j.bcp.2014.08.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/27/2014] [Indexed: 12/12/2022]
Abstract
Increasing evidence indicates that cancer cells rewire their metabolism during tumorigenesis. The high intracellular levels of lactate and reactive oxygen species (ROS) generated during enhanced aerobic glycolysis and mitochondrial oxidative phosphorylation respectively led to oxidative stress. The detoxification of these accumulating metabolites and the equilibrium between reduced and oxidized nicotine adenine dinucleotide (NADH and NAD(+)) are two prominent mechanisms regulating redox status and hence energy homeostasis in tumors. Targeting both processes may thus be selectively cytotoxic for cancer cells. In this context, the impact of poly(ADP-ribose) polymerase (PARP) inhibitors, a class of anticancer agents employed for the treatment of DNA repair deficient tumors, on energy homeostasis and mitochondrial respiration regulation has potential clinical implications. Here we provide an overview of the metabolic reprogramming occurring in cancer cells and discuss the translational perspectives of targeting tumor metabolism and redox balance for antineoplastic therapy.
Collapse
Affiliation(s)
- Judith Michels
- Institut Gustave Roussy, 94805 Villejuif, France; Equipe 11, Centre de Recherche des Cordeliers, Paris 75005, France; Metabolomics Platform, Institut Gustave Roussy, 94805 Villejuif, France
| | - Florine Obrist
- Equipe 11, Centre de Recherche des Cordeliers, Paris 75005, France; Metabolomics Platform, Institut Gustave Roussy, 94805 Villejuif, France; Université de Paris Sud, Paris 11, 94805 Villejuif, France
| | - Maria Castedo
- Equipe 11, Centre de Recherche des Cordeliers, Paris 75005, France; Metabolomics Platform, Institut Gustave Roussy, 94805 Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute, 00144 Roma, Italy.
| | - Guido Kroemer
- Institut Gustave Roussy, 94805 Villejuif, France; Equipe 11, Centre de Recherche des Cordeliers, Paris 75005, France; Metabolomics Platform, Institut Gustave Roussy, 94805 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris 75908, France; Université Paris Descartes, Faculty of Medicine, Paris 75005, France.
| |
Collapse
|
15
|
Brockmann A, Strittmatter T, May S, Stemmer K, Marx A, Brunner T. Structure-function relationship of thiazolide-induced apoptosis in colorectal tumor cells. ACS Chem Biol 2014; 9:1520-7. [PMID: 24810314 DOI: 10.1021/cb500209a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Thiazolides are a novel class of anti-infectious agents against intestinal intracellular and extracellular protozoan parasites, bacteria, and viruses. While the parent compound nitazoxanide (NTZ; 2-(acetolyloxy)-N-(5-nitro-2-thiazolyl)benzamide) has potent antimicrobial activity, the bromo-thiazolide RM4819 (N-(5-bromothiazol-2-yl)-2-hydroxy-3-methylbenzamide) shows only reduced activity. Interestingly, both molecules are able to induce cell death in colon carcinoma cell lines, indicating that the molecular target in intestinal pathogens and in colon cancer cells is different. The detoxification enzyme glutathione S-transferase of class Pi 1 (GSTP1) is frequently overexpressed in various tumors, including colon carcinomas, and limits the efficacy of antitumor chemotherapeutic drugs due to its detoxifying activities. In colorectal tumor cells RM4819 has been shown to interact with GSTP1, and GSTP1 enzymatic activity is required for thiazolide-induced apoptosis. At present it is unclear which molecular structures of RM4819 are required to interact with GSTP1 and to induce cell death in colon carcinoma cell lines. Here, we demonstrate that novel thiazolide derivatives with variation in their substituents of the benzene ring do not significantly affect apoptosis induction in Caco-2 cells, whereas removal of the bromide atom on the thiazole ring leads to a strong reduction of cell death induction in colon cancer cells. We further show that active thiazolides require caspase activation and GSTP1 expression in order to induce apoptosis. We demonstrate that increased glutathione (GSH) levels sensitize colon cancer cells to thiazolides, indicating that both GSTP1 enzymatic activity as well as GSH levels are critical factors in thiazolide-induced cell death.
Collapse
Affiliation(s)
| | | | | | - Kerstin Stemmer
- Helmholtz
Zentrum
Munich, Ingoldstädter Landstr.
1, 85764 Neuherberg, Germany
| | | | | |
Collapse
|
16
|
Ex vivo culture of intestinal crypt organoids as a model system for assessing cell death induction in intestinal epithelial cells and enteropathy. Cell Death Dis 2014; 5:e1228. [PMID: 24832600 PMCID: PMC4047863 DOI: 10.1038/cddis.2014.183] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 12/12/2022]
Abstract
Intestinal epithelial cells (IECs) not only have a critical function in the absorption of nutrients, but also act as a physical barrier between our body and the outside world. Damage and death of the epithelial cells lead to the breakdown of this barrier function and inflammation due to access of the immune system to compounds of the intestinal flora. Intestinal epithelial damage is frequently associated with various inflammatory disorders, chemo- and radiotherapy as well as drug-mediated toxicity. Until recently, intestinal epithelial-damaging activities of drugs and treatments could be tested only in vivo in animal models because of the poor survival rate of primary IECs ex vivo. The three-dimensional culture and outgrowth of intestinal crypt stem cells into organoids have offered new possibilities to culture and study IECs ex vivo. Here we demonstrate that intestinal organoids are a useful and physiologically relevant model system to study cell death and survival in IECs. We further describe a number of microscopy-based as well as colorimetric methods to monitor and score survival and death of intestinal organoids. Finally, the comparison of organoids isolated from gene-deficient mice and wild-type mice allows investigating the role of specific genes in the regulation of IEC death. Owing to their comparable structure and behavior, intestinal organoids may serve as an interesting and physiologically relevant surrogate system for large- and mid-scale in vitro testing of intestinal epithelium-damaging drugs and toxins, and for the investigation of cell death pathways.
Collapse
|
17
|
Strittmatter T, Brockmann A, Pott M, Hantusch A, Brunner T, Marx A. Expanding the scope of human DNA polymerase λ and β inhibitors. ACS Chem Biol 2014; 9:282-90. [PMID: 24171552 DOI: 10.1021/cb4007562] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The exact biological functions of individual DNA polymerases still await clarification, and therefore appropriate reagents to probe their respective functions are required. In the present study, we report the development of a highly potent series of human DNA polymerase λ and β (pol λ and β) inhibitors based on the rhodanine scaffold. Both enzymes are involved in DNA repair and are thus considered as future drug targets. We expanded the chemical diversity of the small-molecule inhibitors arising from a high content screening and designed and synthesized 30 novel analogues. By biochemical evaluation, we discovered 23 highly active compounds against pol λ. Importantly, 10 of these small-molecules selectively inhibited pol λ and not the homologous pol β. We discovered 14 small-molecules that target pol β and found out that they are more potent than known inhibitors. We also investigated whether the discovered compounds sensitize cancer cells toward DNA-damaging reagents. Thus, we cotreated human colorectal cancer cells (Caco-2) with the small-molecule inhibitors and hydrogen peroxide or the approved drug temozolomide. Interestingly, the tested compounds sensitized Caco-2 cells to both genotoxic agents in a DNA repair pathway-dependent manner.
Collapse
Affiliation(s)
- Tobias Strittmatter
- Departments of Chemistry
and Biology, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
| | - Anette Brockmann
- Departments of Chemistry
and Biology, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
| | - Moritz Pott
- Departments of Chemistry
and Biology, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
| | - Annika Hantusch
- Departments of Chemistry
and Biology, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
| | - Thomas Brunner
- Departments of Chemistry
and Biology, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
| | - Andreas Marx
- Departments of Chemistry
and Biology, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
| |
Collapse
|
18
|
Arnold M, Lang E, Modicano P, Bissinger R, Faggio C, Abed M, Lang F. Effect of nitazoxanide on erythrocytes. Basic Clin Pharmacol Toxicol 2013; 114:421-6. [PMID: 24215285 DOI: 10.1111/bcpt.12171] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 10/31/2013] [Indexed: 12/15/2022]
Abstract
Nitazoxanide, a drug effective against a variety of pathogens, triggers apoptosis and is thus considered to be employed against malignancy. Similar to nucleated cells, erythrocytes may undergo an apoptosis-like suicidal cell death or eryptosis. Hallmarks of eryptosis include cell shrinkage and phospholipid scrambling of the cell membrane with translocation of phosphatidylserine to the erythrocyte surface. Stimulators of eryptosis include increase in cytosolic Ca(2+) -activity ([Ca(2+) ]i ). The Ca(2+) -sensitivity of eryptosis is increased by ceramide. This study explored whether nitazoxanide triggers eryptosis. [Ca(2+) ]i was estimated from Fluo3-fluorescence, cell volume from forward scatter, phosphatidylserine exposure from annexin-V-binding, ceramide abundance utilizing fluorescent antibodies and haemolysis from haemoglobin release. A 48-hr exposure to nitazoxanide (1-50 μg/ml) did not significantly modify [Ca(2+) ]i but significantly increased ceramide formation, decreased forward scatter (≥10 μg/ml), increased the percentage of annexin-V-binding erythrocytes (≥10 μg/ml) and, at higher concentrations (≥20 μg/ml), stimulated haemolysis. The stimulation of annexin-V-binding was significantly blunted in the absence of calcium. Nitazoxanide thus stimulates eryptosis, an effect in part due to ceramide formation.
Collapse
Affiliation(s)
- Markus Arnold
- Department of Physiology, Eberhard-Karls-University of Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | |
Collapse
|
19
|
van Huis-Tanja L, Kweekel D, Gelderblom H, Koopman M, Punt K, Guchelaar HJ, van der Straaten T. Concordance of genotype for polymorphisms in DNA isolated from peripheral blood and colorectal cancer tumor samples. Pharmacogenomics 2013; 14:2005-12. [DOI: 10.2217/pgs.13.169] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background & aim: Results from different pharmacogenetic association studies in colorectal cancer are often conflicting. Both peripheral blood and formalin-fixed, paraffin-embedded (FFPE) tissue are routinely used as DNA source. This could cause bias due to somatic alterations in tumor tissue, such as loss of heterozygosity. We therefore compared genotypes in DNA from peripheral blood and FFPE colorectal tumor samples for SNPs with putative influence on the cytotoxicity of chemotherapy. Materials & methods: Eleven SNPs in nine genes involved in anticancer drug metabolism or efficacy were determined in matched samples from blood and FFPE tissue of colorectal tumors by pyrosequencing and TaqMan® techniques. The κ-statistic was calculated to assess concordance. Results: A total of 149 paired FFPE tissue and EDTA blood DNA samples were available for comparison. Overall, 20 out of 1418 genotypes were discordant (1.4%); in ten cases, loss of heterozygosity could not be ruled out. Only GSTP1 showed significant discordance between FFPE tissue and blood genotype (κ = 0.947; 95% CI: 0.896–0.998). Conclusion: FFPE tissue-derived DNA can be used as a valid proxy for germline DNA for a selection of SNPs in (retrospective) pharmacogenetic association studies in colorectal cancer. However, for future studies, genotyping of blood-derived DNA is preferred. Original submitted 29 May 2013; Revision submitted 23 August 2013
Collapse
Affiliation(s)
- Lieke van Huis-Tanja
- Department of Clinical Oncology (K-1-P), Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Dinemarie Kweekel
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Hans Gelderblom
- Department of Clinical Oncology (K-1-P), Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, PO Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Kees Punt
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, PO Box 22660, 1100 DD, Amsterdam, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Tahar van der Straaten
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
20
|
Musdal Y, Hegazy UM, Aksoy Y, Mannervik B. FDA-approved drugs and other compounds tested as inhibitors of human glutathione transferase P1-1. Chem Biol Interact 2013; 205:53-62. [DOI: 10.1016/j.cbi.2013.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/25/2013] [Accepted: 06/03/2013] [Indexed: 11/29/2022]
|
21
|
Hearn JM, Romero-Canelón I, Qamar B, Liu Z, Hands-Portman I, Sadler PJ. Organometallic Iridium(III) anticancer complexes with new mechanisms of action: NCI-60 screening, mitochondrial targeting, and apoptosis. ACS Chem Biol 2013; 8:1335-43. [PMID: 23618382 PMCID: PMC3691721 DOI: 10.1021/cb400070a] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Platinum complexes related to cisplatin, cis-[PtCl2(NH3)2], are successful anticancer drugs; however, other transition metal complexes offer potential for combating cisplatin resistance, decreasing side effects, and widening the spectrum of activity. Organometallic half-sandwich iridium (Ir(III)) complexes [Ir(Cp(x))(XY)Cl](+/0) (Cp(x) = biphenyltetramethylcyclopentadienyl and XY = phenanthroline (1), bipyridine (2), or phenylpyridine (3)) all hydrolyze rapidly, forming monofunctional G adducts on DNA with additional intercalation of the phenyl substituents on the Cp(x) ring. In comparison, highly potent complex 4 (Cp(x) = phenyltetramethylcyclopentadienyl and XY = N,N-dimethylphenylazopyridine) does not hydrolyze. All show higher potency toward A2780 human ovarian cancer cells compared to cisplatin, with 1, 3, and 4 also demonstrating higher potency in the National Cancer Institute (NCI) NCI-60 cell-line screen. Use of the NCI COMPARE algorithm (which predicts mechanisms of action (MoAs) for emerging anticancer compounds by correlating NCI-60 patterns of sensitivity) shows that the MoA of these Ir(III) complexes has no correlation to cisplatin (or oxaliplatin), with 3 and 4 emerging as particularly novel compounds. Those findings by COMPARE were experimentally probed by transmission electron microscopy (TEM) of A2780 cells exposed to 1, showing mitochondrial swelling and activation of apoptosis after 24 h. Significant changes in mitochondrial membrane polarization were detected by flow cytometry, and the potency of the complexes was enhanced ca. 5× by co-administration with a low concentration (5 μM) of the γ-glutamyl cysteine synthetase inhibitor L-buthionine sulfoximine (L-BSO). These studies reveal potential polypharmacology of organometallic Ir(III) complexes, with MoA and cell selectivity governed by structural changes in the chelating ligands.
Collapse
Affiliation(s)
- Jessica M. Hearn
- Warwick
Systems Biology Centre, ‡Department of Chemistry, and §School of Life Sciences, University of Warwick, Gibbet Hill Road,
Coventry CV4 7AL, United Kingdom
| | - Isolda Romero-Canelón
- Warwick
Systems Biology Centre, ‡Department of Chemistry, and §School of Life Sciences, University of Warwick, Gibbet Hill Road,
Coventry CV4 7AL, United Kingdom
| | - Bushra Qamar
- Warwick
Systems Biology Centre, ‡Department of Chemistry, and §School of Life Sciences, University of Warwick, Gibbet Hill Road,
Coventry CV4 7AL, United Kingdom
| | - Zhe Liu
- Warwick
Systems Biology Centre, ‡Department of Chemistry, and §School of Life Sciences, University of Warwick, Gibbet Hill Road,
Coventry CV4 7AL, United Kingdom
| | - Ian Hands-Portman
- Warwick
Systems Biology Centre, ‡Department of Chemistry, and §School of Life Sciences, University of Warwick, Gibbet Hill Road,
Coventry CV4 7AL, United Kingdom
| | - Peter J. Sadler
- Warwick
Systems Biology Centre, ‡Department of Chemistry, and §School of Life Sciences, University of Warwick, Gibbet Hill Road,
Coventry CV4 7AL, United Kingdom
| |
Collapse
|
22
|
Activity of halogeno-thiazolides against Cryptosporidium parvum in experimentally infected immunosuppressed gerbils (Meriones unguiculatus). Antimicrob Agents Chemother 2013; 57:2821-3. [PMID: 23478972 DOI: 10.1128/aac.01538-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nitazoxanide and three halogeno-thiazolides, RM-4850, RM-4865, and RM-5038, were tested against Cryptosporidium parvum in experimentally infected immunosuppressed Mongolian gerbils. Daily 400-mg/kg doses of the four test drugs for 5 to 8 consecutive days produced similar reductions of oocyst shedding. Using early-infected gerbils, a shorter 4-day treatment with RM-5038 reduced oocyst shedding by 95%, compared to 47% for nitazoxanide (P = 0.02), suggesting that RM-5038 is more effective than nitazoxanide under the experimental conditions used.
Collapse
|
23
|
Jones C, St-Jean S, Fréchette I, Bergeron D, Rivard N, Boudreau F. Identification of a novel promyelocytic leukemia zinc-finger isoform required for colorectal cancer cell growth and survival. Int J Cancer 2013; 133:58-66. [PMID: 23280881 DOI: 10.1002/ijc.28008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 10/28/2012] [Accepted: 12/17/2012] [Indexed: 11/06/2022]
Abstract
Promyelocytic leukemia zinc-finger (PLZF) is a transcriptional repressor that regulates proliferation, differentiation and apoptosis among various cellular origins. PLZF expression is upregulated in colorectal cancer cell lines but its putative functional role in this context is unknown. Here, we report the identification of a novel p65 PLZF isoform that results from the usage of an evolutionarily conserved alternative translational initiation site. This isoform is devoid of the classical BTB/POZ domain required for nuclear localization and transcriptional repression. Depletion of p65 PLZF expression in colorectal cancer cell lines results in reduction of cell growth, loss of cell anchorage and increase in cell apoptosis. Overall, these results indicate that p65 PLZF is crucial to maintain colorectal cancer cell adhesion as well as survival and must occur independently of the traditionally viewed transcriptional role of PLZF in the course of these biological processes.
Collapse
Affiliation(s)
- Christine Jones
- Département d'anatomie et biologie cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | | | | | |
Collapse
|
24
|
Müller J, Hemphill A. New approaches for the identification of drug targets in protozoan parasites. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:359-401. [PMID: 23317822 DOI: 10.1016/b978-0-12-407704-1.00007-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Antiparasitic chemotherapy is an important issue for drug development. Traditionally, novel compounds with antiprotozoan activities have been identified by screening of compound libraries in high-throughput systems. More recently developed approaches employ target-based drug design supported by genomics and proteomics of protozoan parasites. In this chapter, the drug targets in protozoan parasites are reviewed. The gene-expression machinery has been among the first targets for antiparasitic drugs and is still under investigation as a target for novel compounds. Other targets include cytoskeletal proteins, proteins involved in intracellular signaling, membranes, and enzymes participating in intermediary metabolism. In apicomplexan parasites, the apicoplast is a suitable target for established and novel drugs. Some drugs act on multiple subcellular targets. Drugs with nitro groups generate free radicals under anaerobic growth conditions, and drugs with peroxide groups generate radicals under aerobic growth conditions, both affecting multiple cellular pathways. Mefloquine and thiazolides are presented as examples for antiprotozoan compounds with multiple (side) effects. The classic approach of drug discovery employing high-throughput physiological screenings followed by identification of drug targets has yielded the mainstream of current antiprotozoal drugs. Target-based drug design supported by genomics and proteomics of protozoan parasites has not produced any antiparasitic drug so far. The reason for this is discussed and a synthesis of both methods is proposed.
Collapse
Affiliation(s)
- Joachim Müller
- Institute of Parasitology, University of Berne, Berne, Switzerland.
| | | |
Collapse
|
25
|
Aituov B, Duisembekova A, Bulenova A, Alibek K. Pathogen-driven gastrointestinal cancers: Time for a change in treatment paradigm? Infect Agent Cancer 2012; 7:18. [PMID: 22873119 PMCID: PMC3508868 DOI: 10.1186/1750-9378-7-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 07/27/2012] [Indexed: 02/06/2023] Open
Abstract
The regulation of cancerous tumor development is converged upon by multiple pathways and factors. Besides environmental factors, gastrointestinal (GI) tract cancer can be caused by chronic inflammation, which is generally induced by bacteria, viruses, and parasites. The role of these inducers in cancer development, cell differentiation and transformation, cell cycle deregulation, and in the expression of tumor-associated genes cannot be ignored. Although Helicobacter pylori activates many oncogenic pathways, particularly those in gastric and colorectal cancers, the role of viruses in tumor development is also significant. Viruses possess significant oncogenic potential to interfere with normal cell cycle control and genome stability, stimulating the growth of deregulated cells. An increasing amount of recent data also implies the association of GI cancers with bacterial colonization and viruses. This review focuses on host-cell interactions that facilitate primary mechanisms of tumorigenesis and provides new insights into novel GI cancer treatments.
Collapse
Affiliation(s)
- Bauyrzhan Aituov
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
| | - Assem Duisembekova
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
| | - Assel Bulenova
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
| | - Kenneth Alibek
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
- Republican Scientific Center for Emergency Care, 3 Kerey and Zhanibek Khan Street, Astana 010000, Kazakhstan
| |
Collapse
|