1
|
Korbecki J, Bosiacki M, Kupnicka P, Barczak K, Ziętek P, Chlubek D, Baranowska-Bosiacka I. Choline kinases: Enzymatic activity, involvement in cancer and other diseases, inhibitors. Int J Cancer 2025; 156:1314-1325. [PMID: 39660774 DOI: 10.1002/ijc.35286] [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: 06/19/2024] [Revised: 10/22/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
One of the aspects of tumor metabolism that distinguish it from healthy tissue is the phosphorylation of choline by choline kinases, which initiates the synthesis of phosphatidylcholine. Presently, there is a lack of comprehensive reviews discussing the current understanding of the role of choline kinase in cancer processes, as well as studies on the anti-tumor properties of choline kinase inhibitors. To address these gaps, this review delves into the enzymatic and non-enzymatic properties of CHKα and CHKβ and explores their precise involvement in cancer processes, particularly cancer cell proliferation. Additionally, we discuss clinical aspects of choline kinases in various tumor types, including pancreatic ductal adenocarcinoma, ovarian cancer, lung adenocarcinoma, lymphoma, leukemia, hepatocellular carcinoma, colon adenocarcinoma, and breast cancer. We examine the potential of CHKα inhibitors as anti-tumor drugs, although they are not yet in the clinical trial phase. Finally, the paper also touches upon the significance of choline kinases in non-cancerous diseases.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Szczecin, Poland
| | - Paweł Ziętek
- Department of Orthopaedics, Traumatology and Orthopaedic Oncology, Pomeranian Medical University, Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
2
|
Bauer LG, Ward JA, Díaz‐Sáez L, Sundström Y, Tolvanen T, Alarcón Barrera JC, Kostidis S, Rogers CM, Panagakou I, Singh U, Rothweiler EM, Gonzalez Orta A, Kaniskan HÜ, Hu J, Jin J, Sievers S, Waldmann H, Giera M, Sundström M, Berg L, Huber KVM. Phenomics-Based Discovery of Novel Orthosteric Choline Kinase Inhibitors. Angew Chem Int Ed Engl 2025; 64:e202420149. [PMID: 39740997 PMCID: PMC11811597 DOI: 10.1002/anie.202420149] [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: 10/18/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025]
Abstract
Choline kinase alpha (CHKA) is a central mediator of cell metabolism linked to cancer and immune regulation. Cellular and clinical evaluation of CHKA has been hampered by challenges in the development of drug-like choline kinase inhibitors. Here, we identify CHKA as an unexpected off-target of histone methyltransferase inhibitors using an integrated phenomic approach. We confirm CHKA as a direct protein target of the aminoquinazolines UNC0638 and UNC0737 using a combination of chemoproteomic, biochemical, cellular, and metabolic profiling assays, possibly explaining the previously reported discrepancies observed for different G9a/GLP inhibitor scaffolds in cellular assays. Using primary human cell model systems, we discover that CHKA modulation impairs IgG secretion and B-cell maturation consistent with the notion that choline metabolism plays an important role in immune signalling. Co-crystal structures of UNC0638 and UNC0737 with CHKA unravel an unexpected binding mode and suggest the inhibitors as attractive starting points for the development of selective chemical tools to further explore the biological role of CHKA in cancer and immune metabolism.
Collapse
Affiliation(s)
- Ludwig G. Bauer
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Jennifer A. Ward
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Laura Díaz‐Sáez
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Yvonne Sundström
- Structural Genomics ConsortiumKarolinska Institutet17176SolnaSweden
- Department of Medicine SolnaKarolinska Institutet and Karolinska University Hospital17176StockholmSweden
| | - Tuomas Tolvanen
- Structural Genomics ConsortiumKarolinska Institutet17176SolnaSweden
- Pelago Bioscience AB17165SolnaSweden
| | | | - Sarantos Kostidis
- Center for Proteomics and MetabolomicsLeiden University Medical Center2333ZALeidenThe Netherlands
| | - Catherine M. Rogers
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Ioanna Panagakou
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Usha Singh
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Elisabeth M. Rothweiler
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Alejandro Gonzalez Orta
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - H. Ümit Kaniskan
- Mount Sinai Center for Therapeutics DiscoveryDepartments of Pharmacological SciencesOncological Sciences and NeuroscienceTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNY 10029USA
| | - Jianping Hu
- Mount Sinai Center for Therapeutics DiscoveryDepartments of Pharmacological SciencesOncological Sciences and NeuroscienceTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNY 10029USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics DiscoveryDepartments of Pharmacological SciencesOncological Sciences and NeuroscienceTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNY 10029USA
| | - Sonja Sievers
- Compound Management and Screening Center at the Max Planck Institute of Molecular Physiology44227DortmundGermany
| | - Herbert Waldmann
- Department of Chemical BiologyMax Planck Institute of Molecular Physiology44227DortmundGermany
| | - Martin Giera
- Center for Proteomics and MetabolomicsLeiden University Medical Center2333ZALeidenThe Netherlands
| | - Michael Sundström
- Structural Genomics ConsortiumKarolinska Institutet17176SolnaSweden
- Department of Medicine SolnaKarolinska Institutet and Karolinska University Hospital17176StockholmSweden
| | - Louise Berg
- Structural Genomics ConsortiumKarolinska Institutet17176SolnaSweden
- Department of Medicine SolnaKarolinska Institutet and Karolinska University Hospital17176StockholmSweden
| | - Kilian V. M. Huber
- Centre for Medicines DiscoveryNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| |
Collapse
|
3
|
Liu J, Jiang B, Xu W, Liu Q, Huang H, Chang X, Ma G, Xu X, Zhou L, Xiao GG, Guo J. Targeted inhibition of CHKα and mTOR in models of pancreatic ductal adenocarcinoma: A novel regimen for metastasis. Cancer Lett 2024; 605:217280. [PMID: 39343354 DOI: 10.1016/j.canlet.2024.217280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic malignancy for which there are currently no effective anti-metastatic therapies. Herein, we employed single-cell RNA sequencing and metabolomics analysis to demonstrate that metastatic cells highly express focal adhesion kinase (FAK), which promotes metastasis by remodeling choline kinase α (CHKα)-dependent choline metabolism. We designed a novel CHKα inhibitor, CHKI-03, and verified its efficacy in inhibiting metastasis in multiple preclinical models. Classical and newly synthesized small-molecule inhibitors have previously been used to assess the therapeutic potential of targeting mTOR and CHKα in various animal models. Mechanistically, FAK activated mTOR and its downstream HIF-1α, thereby elevating CHKα expression and promoting the proliferation, migration, and invasion of PDAC cells, as well as tumor growth and metastasis. Consistently, high expression levels of both FAK and CHKα are correlated with poor prognosis in patients with PDAC. Notably, CHK1-03 inhibited CHKα expression and also suppressed mTORC1 phosphorylation, disrupting the mTORC1-CHKα positive feedback loop. In addition, the combination of CHKI-03 and the mTORC1 inhibitor rapamycin synergistically inhibited tumor growth and metastasis in PDX models. The combination of CHKI-03 and rapamycin demonstrates considerable therapeutic efficacy in PDO models resistant to gemcitabine. Our findings reveal a pivotal mechanism underlying PDAC metastasis regulated by mTORC1-CHKα loop-dependent choline metabolism reprogramming, highlighting the therapeutic potential of this novel regimen for treating PDAC metastasis.
Collapse
Affiliation(s)
- Jianzhou Liu
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Bolun Jiang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 31003, China
| | - Wenchao Xu
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiaofei Liu
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Haoran Huang
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guoxu Ma
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Xudong Xu
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Li Zhou
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Gary Guishan Xiao
- Functional Genomics and Proteomics Center, Creighton University Medical Center, 601N 30th ST, Omaha, NE, 68131, USA
| | - Junchao Guo
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
4
|
Huang T, Chai X, Li S, Liu B, Zhan J, Wang X, Xiao X, Zhu Q, Liu C, Zeng D, Jiang B, Zhou X, He L, Gong Z, Liu M, Zhang X. Rapid Targeted Screening and Identification of Active Ingredients in Herbal Extracts through Ligand-Detected NMR and Database Matching. Anal Chem 2024. [PMID: 39263786 DOI: 10.1021/acs.analchem.4c02255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Herbal extracts are rich sources of active compounds that can be used for drug screening due to their diverse and unique chemical structures. However, traditional methods for screening these compounds are notably laborious and time-consuming. In this manuscript, we introduce a new high-throughput approach that combines nuclear magnetic resonance (NMR) spectroscopy with a tailored database and algorithm to rapidly identify bioactive components in herbal extracts. This method distinguishes characteristic signals and structural motifs of active constituents in the raw extracts through a relaxation-weighted technique, particularly utilizing the perfect echo Carr-Purcell-Meiboom-Gill (peCPMG) sequence, complemented by precise 2D spectroscopic strategies. The cornerstone of our approach is a customized database designed to filter potential compounds based on defined parameters, such as the presence of CHn segments and unique chemical shifts, thereby expediting the identification of promising compounds. This innovative technique was applied to identifying substances interacting with choline kinase α (ChoKα1), resulting in the discovery of four new inhibitors. Our findings demonstrate a powerful tool for unraveling the complex chemical landscape of herbal extracts, considerably facilitating the search for new pharmaceutical candidates. This approach offers an efficient alternative to traditional methods in the quest for drug discovery from natural sources.
Collapse
Affiliation(s)
- Tao Huang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xin Chai
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Shuangli Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Biao Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430071, China
| | - Jianhua Zhan
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xiaohua Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xiong Xiao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qinjun Zhu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Caixiang Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Danyun Zeng
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Jiang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430071, China
- Optics Valley Laboratory, Wuhan 430074, China
| | - Xin Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430071, China
- Optics Valley Laboratory, Wuhan 430074, China
| | - Lichun He
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhou Gong
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Maili Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430071, China
- Optics Valley Laboratory, Wuhan 430074, China
| | - Xu Zhang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement of Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430071, China
- Optics Valley Laboratory, Wuhan 430074, China
| |
Collapse
|
5
|
Deborne J, Benkhaled I, Bouchaud V, Pinaud N, Crémillieux Y. Implantable theranostic device for in vivo real-time NMR evaluation of drug impact in brain tumors. Sci Rep 2024; 14:4541. [PMID: 38402370 PMCID: PMC10894190 DOI: 10.1038/s41598-024-55269-1] [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: 11/01/2023] [Accepted: 02/21/2024] [Indexed: 02/26/2024] Open
Abstract
The evaluation of the efficacy of a drug is a fundamental step in the development of new treatments or in personalized therapeutic strategies and patient management. Ideally, this evaluation should be rapid, possibly in real time, easy to perform and reliable. In addition, it should be associated with as few adverse effects as possible for the patient. In this study, we present a device designed to meet these goals for assessing therapeutic response. This theranostic device is based on the use of magnetic resonance imaging and spectroscopy for the diagnostic aspect and on the application of the convection-enhanced delivery technique for the therapeutic aspect. The miniaturized device is implantable and can be used in vivo in a target tissue. In this study, the device was applied to rodent glioma models with local administration of choline kinase inhibitor and acquisition of magnetic resonance images and spectra at 7 Tesla. The variations in the concentration of key metabolites measured by the device during the administration of the molecules demonstrate the relevance of the approach and the potential of the device.
Collapse
Affiliation(s)
- Justine Deborne
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR 5255, Bordeaux, France
| | - Imad Benkhaled
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR 5255, Bordeaux, France
| | - Véronique Bouchaud
- Centre de Résonance Magnétique des Systèmes Biologiques, Université de Bordeaux, UMR 5536, Bordeaux, France
| | - Noël Pinaud
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR 5255, Bordeaux, France
| | - Yannick Crémillieux
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR 5255, Bordeaux, France.
| |
Collapse
|
6
|
Ghorbani P, Kim SY, Smith TKT, Minarrieta L, Robert-Gostlin V, Kilgour MK, Ilijevska M, Alecu I, Snider SA, Margison KD, Nunes JRC, Woo D, Pember C, O’Dwyer C, Ouellette J, Kotchetkov P, St-Pierre J, Bennett SAL, Lacoste B, Blais A, Nair MG, Fullerton MD. Choline metabolism underpins macrophage IL-4 polarization and RELMα up-regulation in helminth infection. PLoS Pathog 2023; 19:e1011658. [PMID: 37747879 PMCID: PMC10553840 DOI: 10.1371/journal.ppat.1011658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 10/05/2023] [Accepted: 09/05/2023] [Indexed: 09/27/2023] Open
Abstract
Type 2 cytokines like IL-4 are hallmarks of helminth infection and activate macrophages to limit immunopathology and mediate helminth clearance. In addition to cytokines, nutrients and metabolites critically influence macrophage polarization. Choline is an essential nutrient known to support normal macrophage responses to lipopolysaccharide; however, its function in macrophages polarized by type 2 cytokines is unknown. Using murine IL-4-polarized macrophages, targeted lipidomics revealed significantly elevated levels of phosphatidylcholine, with select changes to other choline-containing lipid species. These changes were supported by the coordinated up-regulation of choline transport compared to naïve macrophages. Pharmacological inhibition of choline metabolism significantly suppressed several mitochondrial transcripts and dramatically inhibited select IL-4-responsive transcripts, most notably, Retnla. We further confirmed that blocking choline metabolism diminished IL-4-induced RELMα (encoded by Retnla) protein content and secretion and caused a dramatic reprogramming toward glycolytic metabolism. To better understand the physiological implications of these observations, naïve or mice infected with the intestinal helminth Heligmosomoides polygyrus were treated with the choline kinase α inhibitor, RSM-932A, to limit choline metabolism in vivo. Pharmacological inhibition of choline metabolism lowered RELMα expression across cell-types and tissues and led to the disappearance of peritoneal macrophages and B-1 lymphocytes and an influx of infiltrating monocytes. The impaired macrophage activation was associated with some loss in optimal immunity to H. polygyrus, with increased egg burden. Together, these data demonstrate that choline metabolism is required for macrophage RELMα induction, metabolic programming, and peritoneal immune homeostasis, which could have important implications in the context of other models of infection or cancer immunity.
Collapse
Affiliation(s)
- Peyman Ghorbani
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Sang Yong Kim
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
| | - Tyler K. T. Smith
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Lucía Minarrieta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Victoria Robert-Gostlin
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marisa K. Kilgour
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
| | - Maja Ilijevska
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
| | - Irina Alecu
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Shayne A. Snider
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Kaitlyn D. Margison
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Julia R. C. Nunes
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Daniel Woo
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
| | - Ciara Pember
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
| | - Conor O’Dwyer
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Julie Ouellette
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Pavel Kotchetkov
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Steffany A. L. Bennett
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Brain and Mind Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Brain and Mind Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexandre Blais
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Brain and Mind Institute, University of Ottawa, Ottawa, Ontario, Canada
- Éric Poulin Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Meera G. Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
| | - Morgan D. Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
7
|
Luque-Navarro PM, Carrasco-Jiménez MP, Goracci L, Paredes JM, Espinar-Barranco L, Valverde-Pozo J, Torretta A, Parisini E, Mariotto E, Marchioro C, Laso A, Marco C, Viola G, Lanari D, López Cara LC. New bioisosteric sulphur-containing choline kinase inhibitors with a tracked mode of action. Eur J Med Chem 2023; 246:115003. [PMID: 36493617 DOI: 10.1016/j.ejmech.2022.115003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Since the identification of human choline kinase as a protein target against cancer progression, many compounds have been designed to inhibit its function and reduce the biosynthesis of phosphatidylcholine. Herein, we propose a series of bioisosteric inhibitors that are based on the introduction of sulphur and feature improved activity and lipophilic/hydrophilic balance. The evaluation of the inhibitory and of the antiproliferative properties of the PL (dithioethane) and FP (disulphide) libraries led to the identification of PL 48, PL 55 and PL 69 as the most active compounds of the series. Docking analysis using FLAP suggests that for hits to leads, binding mostly involves an interaction with the Mg2+ cofactor, or its destabilization. The most active compounds of the two series are capable of inducing apoptosis following the mitochondrial pathway and to significantly reduce the expression of anti-apoptotic proteins such as the Mcl-1. The fluorescence properties of the compounds of the PL library allowed the tracking of their mode of action, while PAINS (Pan Assays Interference Structures) filtration databases suggest the lack of any unspecific biological response.
Collapse
Affiliation(s)
- Pilar M Luque-Navarro
- Department of Pharmaceutical and Organic Chemistry, Faculty of Pharmacy, University of Granada, Campus of Cartuja s/n, Granada, 18071, Spain; Department of Pharmaceutical Sciences, University of Perugia, Perugia, 06123 Italy
| | - M Paz Carrasco-Jiménez
- Department of Biochemistry and Molecular Biology I, University of Granada, Campus of Fuentenueva s/n, Granada, 18071, Spain.
| | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, 06123, Italy
| | - Jose M Paredes
- Department of Physical-Chemistry, Faculty of Pharmacy, University of Granada, Campus of Cartuja s/n, Granada, 18071, Spain
| | - Laura Espinar-Barranco
- Department of Physical-Chemistry, Faculty of Pharmacy, University of Granada, Campus of Cartuja s/n, Granada, 18071, Spain
| | - Javier Valverde-Pozo
- Department of Physical-Chemistry, Faculty of Pharmacy, University of Granada, Campus of Cartuja s/n, Granada, 18071, Spain
| | - Archimede Torretta
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Via Pascoli 70/3, Milano, 20133, Italy
| | - Emilio Parisini
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Via Pascoli 70/3, Milano, 20133, Italy; Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia; Department of Chemistry "G. Ciamician", University of Bologna, Via Selmi 2, Bologna, 40126, Italy
| | - Elena Mariotto
- Department of Woman's and Child's Health, Laboratory of Oncohematology, University of Padova, Padova, 35128, Italy
| | - Chiara Marchioro
- Department of Woman's and Child's Health, Laboratory of Oncohematology, University of Padova, Padova, 35128, Italy
| | - Alejandro Laso
- Department of Biochemistry and Molecular Biology I, University of Granada, Campus of Fuentenueva s/n, Granada, 18071, Spain
| | - Carmen Marco
- Department of Biochemistry and Molecular Biology I, University of Granada, Campus of Fuentenueva s/n, Granada, 18071, Spain
| | - Giampietro Viola
- Department of Woman's and Child's Health, Laboratory of Oncohematology, University of Padova, Padova, 35128, Italy; Istituto di Ricerca Pediatrica (IRP) Fondazione Città della Speranza, Corso Stati Uniti 4, Padova, 35128, Italy.
| | - Daniela Lanari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, 06123 Italy.
| | - Luisa Carlota López Cara
- Department of Pharmaceutical and Organic Chemistry, Faculty of Pharmacy, University of Granada, Campus of Cartuja s/n, Granada, 18071, Spain.
| |
Collapse
|
8
|
Molecular and metabolic alterations of 2,3-dihydroquinazolin-4(1H)-one derivatives in prostate cancer cell lines. Sci Rep 2022; 12:21599. [PMID: 36517571 PMCID: PMC9751122 DOI: 10.1038/s41598-022-26148-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PC) is the second most common tumor in males worldwide. The lack of effective medication and the development of multidrug resistance towards current chemotherapeutic agents urge the need to discover novel compounds and therapeutic targets for PC. Herein, seven synthesized 2,3-dihydroquinazolin-4(1H)-one analogues were evaluated for their anticancer activity against PC3 and DU145 cancer cell lines using MTT, scratch-wound healing, adhesion and invasion assays. Besides, a liquid chromatography mass spectrometry (LC-MS)-based metabolomics approach was followed to identify the biochemical pathways altered in DU145 cancer cells upon exposure to dihydroquinazolin derivatives. The seven compounds showed sufficient cytotoxicity and significantly suppressed DU145 and PC3 migration after 48 and 72 h. C2 and C5 had the most potent effect with IC50 < 15 µM and significantly inhibited PC cell adhesion and invasion. Metabolomics revealed that C5 disturbed the level of metabolites involved in essential processes for cancer cell proliferation, progression and growth including energy production, redox homeostasis, amino acids and polyamine metabolisms and choline phospholipid metabolism. The data presented herein highlighted the importance of these compounds as potential anticancer agents particularly C5, and pointed to the promising role of metabolomics as a new analytical approach to investigate the antiproliferative activity of synthesized compounds and identify new therapeutic targets.
Collapse
|
9
|
Miriam Jose A, Rasool M. Choline kinase: An underappreciated rheumatoid arthritis therapeutic target. Life Sci 2022; 309:121031. [DOI: 10.1016/j.lfs.2022.121031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/15/2022]
|
10
|
Lacal JC, Perona R, de Castro J, Cebrián A. Choline Kinase α Inhibitors MN58b and RSM932A Enhances the Antitumor Response to Cisplatin in Lung Tumor Cells. Pharmaceutics 2022; 14:pharmaceutics14061143. [PMID: 35745716 PMCID: PMC9230389 DOI: 10.3390/pharmaceutics14061143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
Lung cancer is one of the main causes of death in developed countries, and non-small cell lung cancer (NSCLC) is the most frequent type (80% of patients). In advanced NSCLC, platinum-based chemotherapy is the frontline palliative treatment, but less than 5% of patients achieve prolonged survival. Immunotherapy has recently been proposed as the standard of care (SoC) as either monotherapy or in combination with chemotherapy for advanced NSCLC. The levels of expression of PD-L1 are the only predictive biomarkers for patient assessment. Although around 30% of patients receiving immunotherapy achieve 5-year survival, a significant number does not benefit from this novel therapeutic approach. Therefore, there is a need for novel strategies to improve clinical outcomes. The expression level of choline kinase α (ChoKα) is increased in a large number of human tumors, including NSCLC tumors, and constitutes an independent prognostic factor for early-stage NSCLC patients. Thus, ChoKα has been postulated as a new target drug in cancer therapy. The combination of cisplatin with novel targeted drugs such as choline kinase inhibitors may improve both the survival rates and the quality of life of NSCLC patients and may serve as the basis for the development of new therapeutic approaches. To that aim, we developed several in vitro and in vivo approaches to assess the antitumor activity of a novel combination regimen using cisplatin and ChoKα inhibitors. Our results suggest that a proper combination of specific inhibitors of the NSCLC prognostic factor ChoKα and platinum-based conventional chemotherapy might constitute a new, efficient treatment approach for NSCLC patients. This novel approach may help reduce the toxicity profile associated with cisplatin since, despite the advances in NSCLC management in recent years, the overall 5-year survival rate is still poor.
Collapse
Affiliation(s)
- Juan Carlos Lacal
- Instituto de Investigaciones Biomédicas, CSIC/UAM, 28029 Madrid, Spain; (R.P.); (A.C.)
- Instituto de Investigación Sanitaria Hospital La Paz, IDIPAZ, 28046 Madrid, Spain;
- Correspondence: ; Tel.: +34-914-975-438
| | - Rosario Perona
- Instituto de Investigaciones Biomédicas, CSIC/UAM, 28029 Madrid, Spain; (R.P.); (A.C.)
- Instituto de Investigación Sanitaria Hospital La Paz, IDIPAZ, 28046 Madrid, Spain;
| | - Javier de Castro
- Instituto de Investigación Sanitaria Hospital La Paz, IDIPAZ, 28046 Madrid, Spain;
| | - Arancha Cebrián
- Instituto de Investigaciones Biomédicas, CSIC/UAM, 28029 Madrid, Spain; (R.P.); (A.C.)
- Instituto de Investigación Sanitara Fundación Jiménez Díaz, 28040 Madrid, Spain
| |
Collapse
|
11
|
Saito RDF, Andrade LNDS, Bustos SO, Chammas R. Phosphatidylcholine-Derived Lipid Mediators: The Crosstalk Between Cancer Cells and Immune Cells. Front Immunol 2022; 13:768606. [PMID: 35250970 PMCID: PMC8889569 DOI: 10.3389/fimmu.2022.768606] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/13/2022] [Indexed: 01/16/2023] Open
Abstract
To become resistant, cancer cells need to activate and maintain molecular defense mechanisms that depend on an energy trade-off between resistance and essential functions. Metabolic reprogramming has been shown to fuel cell growth and contribute to cancer drug resistance. Recently, changes in lipid metabolism have emerged as an important driver of resistance to anticancer agents. In this review, we highlight the role of choline metabolism with a focus on the phosphatidylcholine cycle in the regulation of resistance to therapy. We analyze the contribution of phosphatidylcholine and its metabolites to intracellular processes of cancer cells, both as the major cell membrane constituents and source of energy. We further extended our discussion about the role of phosphatidylcholine-derived lipid mediators in cellular communication between cancer and immune cells within the tumor microenvironment, as well as their pivotal role in the immune regulation of therapeutic failure. Changes in phosphatidylcholine metabolism are part of an adaptive program activated in response to stress conditions that contribute to cancer therapy resistance and open therapeutic opportunities for treating drug-resistant cancers.
Collapse
Affiliation(s)
- Renata de Freitas Saito
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Luciana Nogueira de Sousa Andrade
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Silvina Odete Bustos
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Zhang X, Zhang R, Liu P, Zhang R, Ning J, Ye Y, Yu W, Yu J. ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14030835. [PMID: 35159102 PMCID: PMC8834475 DOI: 10.3390/cancers14030835] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/14/2022] [Accepted: 02/02/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary We found that low expression of ATP8B1 was associated with poor prognosis, and involved in the dysregulation of glutathione (GSH) synthesis and choline metabolism in lung squamous cell carcinoma (LUSC) samples of The Cancer Genome Atlas (TCGA) and Tianjin Medical University Cancer Institute and Hospital (TJMUCH) cohort. We further constructed ATP8B1 knockdown of LUSC cell lines H520SH-ATP8B1 and SK-MES-1SH-ATP8B1 to investigate how ATP8B1 knockdown promoted cell proliferation, migration, and invasion in vitro and in vivo via upregulation of the CHKA-dependent choline metabolism pathway. We identified that ATP8B1 knockdown and CHKA upregulation can lead to mitochondrial dysfunction and high reduction-oxidation (REDOX) homeostasis, which may be involved in the roles of cardiolipin in maintaining mitochondrial dynamics and phospholipid homeostasis. Therefore, we proposed ATP8B1 as a novel predictive biomarker in LUSC and targeting ATP8B1-driven specific metabolic disorder might be a promising therapeutic strategy for LUSC. Abstract The flippase ATPase class I type 8b member 1 (ATP8B1) is essential for maintaining the stability and polarity of the epithelial membrane and can translocate specific phospholipids from the outer membrane to the inner membrane of the cell. Although ATP8B1 plays important roles in cell functions, ATP8B1 has been poorly studied in tumors and its prognostic value in patients with lung squamous cell carcinoma (LUSC) remains unclear. By investigating the whole genomic expression profiles of LUSC samples from The Cancer Genome Atlas (TCGA) database and Tianjin Medical University Cancer Institute and Hospital (TJMUCH) cohort, we found that low expression of ATP8B1 was associated with poor prognosis of LUSC patients. The results from cellular experiments and a xenograft-bearing mice model indicated that ATP8B1 knockdown firstly induced mitochondrial dysfunction and promoted ROS production. Secondly, ATP8B1 knockdown promoted glutathione synthesis via upregulation of the CHKA-dependent choline metabolism pathway, therefore producing and maintaining high-level intracellular REDOX homeostasis to aggravate carcinogenesis and progression of LUSC. In summary, we proposed ATP8B1 as a novel predictive biomarker in LUSC and targeting ATP8B1-driven specific metabolic disorder might be a promising therapeutic strategy for LUSC.
Collapse
Affiliation(s)
- Xiao Zhang
- Cancer Molecular Diagnostics Core, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China; (X.Z.); (R.Z.); (P.L.); (R.Z.); (J.N.); (Y.Y.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- National Clinical Research Center of Caner, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China;
| | - Rui Zhang
- Cancer Molecular Diagnostics Core, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China; (X.Z.); (R.Z.); (P.L.); (R.Z.); (J.N.); (Y.Y.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Pengpeng Liu
- Cancer Molecular Diagnostics Core, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China; (X.Z.); (R.Z.); (P.L.); (R.Z.); (J.N.); (Y.Y.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Runjiao Zhang
- Cancer Molecular Diagnostics Core, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China; (X.Z.); (R.Z.); (P.L.); (R.Z.); (J.N.); (Y.Y.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- National Clinical Research Center of Caner, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China;
| | - Junya Ning
- Cancer Molecular Diagnostics Core, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China; (X.Z.); (R.Z.); (P.L.); (R.Z.); (J.N.); (Y.Y.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- National Clinical Research Center of Caner, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China;
| | - Yingnan Ye
- Cancer Molecular Diagnostics Core, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China; (X.Z.); (R.Z.); (P.L.); (R.Z.); (J.N.); (Y.Y.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wenwen Yu
- National Clinical Research Center of Caner, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China;
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China; (X.Z.); (R.Z.); (P.L.); (R.Z.); (J.N.); (Y.Y.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Correspondence: ; Tel.: +86-22-23340123; Fax: +86-22-23340123 (ext. 6325)
| |
Collapse
|
13
|
Khalifa M, Few LL, Too WCS. Phage-Choline Kinase Inhibitor Combination to Control Pseudomonas aeruginosa: A Promising Combo. Mini Rev Med Chem 2021; 22:1281-1288. [PMID: 34961459 DOI: 10.2174/1389557521666211213160256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/09/2021] [Accepted: 10/25/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Pseudomonas aeruginosa is one of the most prevalent opportunistic pathogens in humans that has thrived and proved to be difficult to control in this "post-antibiotic era." Antibiotic alternatives are necessary for fighting against this resilient bacterium. Even though phages might not be "the wonder drug" that solves everything, they still provide a viable option to combat P. aeruginosa and curb the threat it imposes. MAIN FINDINGS The combination of antibiotics with phages, however, poses a propitious treatment option for P. aeruginosa. Choline kinase (ChoK) is the enzyme that synthesizes phosphorylcholine subsequently incorporated into lipopolysaccharide located at the outer membrane of gram-negative bacteria. Recently, inhibition of ChoKs has been proposed as a promising antibacterial strategy. Successful docking of Hemicholinium-3, a choline kinase inhibitor, to the model structure of P. aeruginosa ChoK also supports the use of this inhibitor or its derivatives to inhibit the growth of this microorganism. CONCLUSION Therefore, the combination of the novel antimicrobial "choline kinase inhibitors (ChoKIs)" with a phage cocktail or synthetic phages as a potential treatment for P. aeruginosa infection has been proposed.
Collapse
Affiliation(s)
- Moad Khalifa
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan. Malaysia
| | - Ling Ling Few
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan. Malaysia
| | - Wei Cun See Too
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan. Malaysia
| |
Collapse
|
14
|
Guido ME, Monjes NM, Wagner PM, Salvador GA. Circadian Regulation and Clock-Controlled Mechanisms of Glycerophospholipid Metabolism from Neuronal Cells and Tissues to Fibroblasts. Mol Neurobiol 2021; 59:326-353. [PMID: 34697790 DOI: 10.1007/s12035-021-02595-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/07/2021] [Indexed: 12/26/2022]
Abstract
Along evolution, living organisms developed a precise timekeeping system, circadian clocks, to adapt life to the 24-h light/dark cycle and temporally regulate physiology and behavior. The transcriptional molecular circadian clock and metabolic/redox oscillator conforming these clocks are present in organs, tissues, and even in individual cells, where they exert circadian control over cellular metabolism. Disruption of the molecular clock may cause metabolic disorders and higher cancer risk. The synthesis and degradation of glycerophospholipids (GPLs) is one of the most highly regulated metabolisms across the 24-h cycle in terms of total lipid content and enzyme expression and activity in the nervous system and individual cells. Lipids play a plethora of roles (membrane biogenesis, energy sourcing, signaling, and the regulation of protein-chromatin interaction, among others), making control of their metabolism a vital checkpoint in the cellular organization of physiology. An increasing body of evidence clearly demonstrates an orchestrated and sequential series of events occurring in GPL metabolism across the 24-h day in diverse retinal cell layers, immortalized fibroblasts, and glioma cells. Moreover, the clock gene Per1 and other circadian-related genes are tightly involved in the regulation of GPL synthesis in quiescent cells. However, under proliferation, the metabolic oscillator continues to control GPL metabolism of brain cancer cells even after molecular circadian clock disruption, reflecting the crucial role of the temporal metabolism organization in cell preservation. The aim of this review is to examine the control exerted by circadian clocks over GPL metabolism, their synthesizing enzyme expression and activities in normal and tumorous cells of the nervous system and in immortalized fibroblasts.
Collapse
Affiliation(s)
- Mario E Guido
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina.
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina.
| | - Natalia M Monjes
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
| | - Paula M Wagner
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
- Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
| | - Gabriela A Salvador
- INIBIBB-UNS-CONICET, Departamento de Biología, Bioquímica y Farmacia, UNS, Bahía Blanca, Argentina
| |
Collapse
|
15
|
Wang N, Brickute D, Braga M, Barnes C, Lu H, Allott L, Aboagye EO. Novel Non-Congeneric Derivatives of the Choline Kinase Alpha Inhibitor ICL-CCIC-0019. Pharmaceutics 2021; 13:1078. [PMID: 34371769 PMCID: PMC8309005 DOI: 10.3390/pharmaceutics13071078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 01/07/2023] Open
Abstract
Choline kinase alpha (CHKA) is a promising target for the development of cancer therapeutics. We have previously reported ICL-CCIC-0019, a potent CHKA inhibitor with high cellular activity but with some unfavorable pharmacological properties. In this work, we present an active analogue of ICL-CCIC-0019 bearing a piperazine handle (CK146) to facilitate further structural elaboration of the pharmacophore and thus improve the biological profile. Two different strategies were evaluated in this study: (1) a prodrug approach whereby selective CHKA inhibition could be achieved through modulating the activity of CK146, via the incorporation of an ε-(Ac) Lys motif, cleavable by elevated levels of histone deacetylase (HDAC) and cathepsin L (CTSL) in tumour cells; (2) a prostate-specific membrane antigen (PSMA) receptor targeted delivery strategy. Prodrug (CK145) and PSMA-targeted (CK147) derivatives were successfully synthesized and evaluated in vitro. While the exploitation of CK146 in those two strategies did not deliver the expected results, important and informative structure-activity relationships were observed and have been reported.
Collapse
Affiliation(s)
- Ning Wang
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (N.W.); (D.B.); (M.B.); (C.B.); (H.L.)
| | - Diana Brickute
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (N.W.); (D.B.); (M.B.); (C.B.); (H.L.)
| | - Marta Braga
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (N.W.); (D.B.); (M.B.); (C.B.); (H.L.)
| | - Chris Barnes
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (N.W.); (D.B.); (M.B.); (C.B.); (H.L.)
| | - Haonan Lu
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (N.W.); (D.B.); (M.B.); (C.B.); (H.L.)
| | - Louis Allott
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (N.W.); (D.B.); (M.B.); (C.B.); (H.L.)
- Positron Emission Tomography Research Centre, Faculty of Health Sciences, University of Hull, Kingston upon Hull HU6 7RX, UK
| | - Eric O. Aboagye
- Comprehensive Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (N.W.); (D.B.); (M.B.); (C.B.); (H.L.)
| |
Collapse
|
16
|
Lacal JC, Zimmerman T, Campos JM. Choline Kinase: An Unexpected Journey for a Precision Medicine Strategy in Human Diseases. Pharmaceutics 2021; 13:788. [PMID: 34070409 PMCID: PMC8226952 DOI: 10.3390/pharmaceutics13060788] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 12/17/2022] Open
Abstract
Choline kinase (ChoK) is a cytosolic enzyme that catalyzes the phosphorylation of choline to form phosphorylcholine (PCho) in the presence of ATP and magnesium. ChoK is required for the synthesis of key membrane phospholipids and is involved in malignant transformation in a large variety of human tumours. Active compounds against ChoK have been identified and proposed as antitumor agents. The ChoK inhibitory and antiproliferative activities of symmetrical bispyridinium and bisquinolinium compounds have been defined using quantitative structure-activity relationships (QSARs) and structural parameters. The design strategy followed in the development of the most active molecules is presented. The selective anticancer activity of these structures is also described. One promising anticancer compound has even entered clinical trials. Recently, ChoKα inhibitors have also been proposed as a novel therapeutic approach against parasites, rheumatoid arthritis, inflammatory processes, and pathogenic bacteria. The evidence for ChoKα as a novel drug target for approaches in precision medicine is discussed.
Collapse
Affiliation(s)
- Juan Carlos Lacal
- Instituto de Investigaciones Biomédicas, CSIC, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz, IDIPAZ, 28046 Madrid, Spain
| | - Tahl Zimmerman
- Food Microbiology and Biotechnology Laboratory, Department of Family and Consumer Sciences, College of Agriculture and Environmental Sciences, North Carolina University, 1601 East Market Street, Greensboro, NC 27411, USA;
| | - Joaquín M. Campos
- Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, c/Campus de Cartuja, s/n, Universidad de Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs. GRANADA), SAS-Universidad de Granada, 18071 Granada, Spain
| |
Collapse
|
17
|
Identification of Monotonically Differentially Expressed Genes across Pathologic Stages for Cancers. JOURNAL OF ONCOLOGY 2020; 2020:8458190. [PMID: 33273919 PMCID: PMC7676961 DOI: 10.1155/2020/8458190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/17/2020] [Accepted: 10/28/2020] [Indexed: 12/09/2022]
Abstract
Given the fact that cancer is a multistage progression process resulting from genetic sequence mutations, the genes whose expression values increase or decrease monotonically across pathologic stages are potentially involved in tumor progression. This may provide insightful clues about how human cancers advance, thereby facilitating more personalized treatments. By replacing the expression values of genes with their GeneRanks, we propose a procedure capable of identifying monotonically differentially expressed genes (MEGs) as the disease advances. Using three real-world gene expression data that cover three distinct cancer types-colon, esophageal, and lung cancers-the proposed procedure has demonstrated excellent performance in detecting the potential MEGs. To conclude, the proposed procedure can detect MEGs across pathologic stages of cancers very efficiently and is thus highly recommended.
Collapse
|
18
|
Song P, Shen X. Juice from Fructus Rosae Roxburghii normalizes blood lipids in mice with diet-induced hyperlipidemia* †. Food Sci Nutr 2020; 8:6069-6082. [PMID: 33282259 PMCID: PMC7684604 DOI: 10.1002/fsn3.1897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 12/18/2022] Open
Abstract
Fructus Rosae Roxburghii (FRR) as a dietary supplement is considered to possess anti-atherosclerosis (AS), and hyperlipidemia (HLP) is material basis for AS formation, so the effects and molecular mechanism of FRR on diet-induced hyperlipidemic mice were explored. In Diet IV2 group, hepatic steatosis was significantly relieved; meanwhile, TC, TG, LDL-C, HDL-C, and ASI in serum were regulated to control level. Thirty-seven DCEG in Diet I, Diet II, and Diet IV2 groups were obtained by RNA-seq analysis. Relative mRNA levels were further determined by qRT-PCR, of which 28 genes were matched with those detected by RNA-seq. Ten DCEP were verified by targeted quantitative proteomic analysis, but expressive patterns of only six proteins were correlated with qRT-PCR data. These DCEG and DCEP played important roles in regulating the biosynthesis of BAs and steroids, fatty acid metabolism, and LPO production. They might cooperatively regulate the function of HDL or RCT by PPAR signaling pathway under the FRR action. As we know, it is the first time the potential anti-atherosclerotic mechanism of FRR regulating the blood lipids was explored.
Collapse
Affiliation(s)
- Pingping Song
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Key Laboratory of Optimal Utilization of Natural Medicine ResourcesSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuiyangChina
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Key Laboratory of Optimal Utilization of Natural Medicine ResourcesSchool of Pharmaceutical SciencesGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
19
|
Rubio-Ruiz B, Serrán-Aguilera L, Hurtado-Guerrero R, Conejo-García A. Recent advances in the design of choline kinase α inhibitors and the molecular basis of their inhibition. Med Res Rev 2020; 41:902-927. [PMID: 33103259 DOI: 10.1002/med.21746] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/25/2020] [Accepted: 10/12/2020] [Indexed: 12/15/2022]
Abstract
Upregulated choline metabolism, characterized by an increase in phosphocholine (PCho), is a hallmark of oncogenesis and tumor progression. Choline kinase (ChoK), the enzyme responsible for PCho synthesis, has consequently become a promising drug target for cancer therapy and as such a significant number of ChoK inhibitors have been developed over the last few decades. More recently, due to the role of this enzyme in other pathologies, ChoK inhibitors have also been used in new therapeutic approaches against malaria and rheumatoid arthritis. Here, we review research results in the field of ChoKα inhibitors from their synthesis to the molecular basis of their binding mode. Strategies for the development of inhibitors and their selectivity on ChoKα over ChoKβ, the plasticity of the choline-binding site, the discovery of new exploitable binding sites, and the allosteric properties of this enzyme are highlighted. The outcomes summarized in this review will be a useful guide to develop new multifunctional potent drugs for the treatment of various human diseases.
Collapse
Affiliation(s)
- Belén Rubio-Ruiz
- Department of Medicinal and Organic Chemistry, Faculty of Pharmacy, University of Granada, Granada, Spain.,Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, Spain
| | - Lucía Serrán-Aguilera
- Department of Medicinal and Organic Chemistry, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Ramón Hurtado-Guerrero
- Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Zaragoza, Spain.,Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark.,Laboratorio de Microscopías Avanzada, University of Zaragoza, Zaragoza, Spain.,ARAID Foundation, Zaragoza, Spain
| | - Ana Conejo-García
- Department of Medicinal and Organic Chemistry, Faculty of Pharmacy, University of Granada, Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, Spain
| |
Collapse
|
20
|
Identification and validation of novel and more effective choline kinase inhibitors against Streptococcus pneumoniae. Sci Rep 2020; 10:15418. [PMID: 32963303 PMCID: PMC7508948 DOI: 10.1038/s41598-020-72165-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/24/2020] [Indexed: 11/08/2022] Open
Abstract
Streptococcus pneumoniae choline kinase (sChoK) has previously been proposed as a drug target, yet the effectiveness of the first and only known inhibitor of sChoK, HC-3, is in the millimolar range. The aim of this study was thus to further validate sChoK as a potential therapeutic target by discovering more powerful sChoK inhibitors. LDH/PK and colorimetric enzymatic assays revealed two promising sChoK inhibitor leads RSM-932A and MN58b that were discovered with IC50 of 0.5 and 150 μM, respectively, and were shown to be 2–4 magnitudes more potent than the previously discovered inhibitor HC-3. Culture assays showed that the minimum inhibitory concentration (MIC) of RSM-932A and MN58b for S. pneumoniae was 0.4 μM and 10 μM, respectively, and the minimum lethal concentration (MLC) was 1.6 μM and 20 μM, respectively. Western blot monitoring of teichoic acid production revealed differential patterns in response to each inhibitor. In addition, both inhibitors possessed a bacteriostatic mechanism of action, and neither interfered with the autolytic effects of vancomycin. Cells treated with MN58b but not RSM-932A were more sensitive to a phosphate induced autolysis with respect to the untreated cells. SEM studies revealed that MN58b distorted the cell wall, a result consistent with the apparent teichoic acid changes. Two novel and more highly potent putative inhibitors of sChoK, MN58b and RSM-932A, were characterized in this study. However, the effects of sChoK inhibitors can vary at the cellular level. sChoK inhibition is a promising avenue to follow in the development of therapeutics for treatment of S. pneumoniae.
Collapse
|
21
|
Khalifa M, Few LL, See Too WC. ChoK-ing the Pathogenic Bacteria: Potential of Human Choline Kinase Inhibitors as Antimicrobial Agents. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1823485. [PMID: 32695809 PMCID: PMC7368946 DOI: 10.1155/2020/1823485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/25/2020] [Accepted: 06/29/2020] [Indexed: 01/12/2023]
Abstract
Novel antimicrobial agents are crucial to combat antibiotic resistance in pathogenic bacteria. Choline kinase (ChoK) in bacteria catalyzes the synthesis of phosphorylcholine, which is subsequently incorporated into the cell wall or outer membrane. In certain species of bacteria, phosphorylcholine is also used to synthesize membrane phosphatidylcholine. Numerous human ChoK inhibitors (ChoKIs) have been synthesized and tested for anticancer properties. Inhibition of S. pneumoniae ChoK by human ChoKIs showed a promising effect by distorting the cell wall and retarded the growth of this pathogen. Comparison of amino acid sequences at the catalytic sites of putative choline kinases from pathogenic bacteria and human enzymes revealed striking sequence conservation that supports the potential application of currently available ChoKIs for inhibiting bacterial enzymes. We also propose the combined use of ChoKIs and nanoparticles for targeted delivery to the pathogen while shielding the human host from any possible side effects of the inhibitors. More research should focus on the verification of putative bacterial ChoK activities and the characterization of ChoKIs with active enzymes. In conclusion, the presence of ChoK in a wide range of pathogenic bacteria and the distinct function of this enzyme has made it an attractive drug target. This review highlighted the possibility of "choking" bacterial ChoKs by using human ChoKIs.
Collapse
Affiliation(s)
- Moad Khalifa
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Ling Ling Few
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Wei Cun See Too
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
22
|
Gokhale S, Lu W, Zhu S, Liu Y, Hart RP, Rabinowitz JD, Xie P. Elevated Choline Kinase α-Mediated Choline Metabolism Supports the Prolonged Survival of TRAF3-Deficient B Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2019; 204:459-471. [PMID: 31826940 DOI: 10.4049/jimmunol.1900658] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/13/2019] [Indexed: 12/27/2022]
Abstract
Specific deletion of the tumor suppressor TRAF3 from B lymphocytes in mice leads to the prolonged survival of mature B cells and expanded B cell compartments in secondary lymphoid organs. In the current study, we investigated the metabolic basis of TRAF3-mediated regulation of B cell survival by employing metabolomic, lipidomic, and transcriptomic analyses. We compared the polar metabolites, lipids, and metabolic enzymes of resting splenic B cells purified from young adult B cell-specific Traf3 -/- and littermate control mice. We found that multiple metabolites, lipids, and enzymes regulated by TRAF3 in B cells are clustered in the choline metabolic pathway. Using stable isotope labeling, we demonstrated that phosphocholine and phosphatidylcholine biosynthesis was markedly elevated in Traf3 -/- mouse B cells and decreased in TRAF3-reconstituted human multiple myeloma cells. Furthermore, pharmacological inhibition of choline kinase α, an enzyme that catalyzes phosphocholine synthesis and was strikingly increased in Traf3 -/- B cells, substantially reversed the survival phenotype of Traf3 -/- B cells both in vitro and in vivo. Taken together, our results indicate that enhanced phosphocholine and phosphatidylcholine synthesis supports the prolonged survival of Traf3 -/- B lymphocytes. Our findings suggest that TRAF3-regulated choline metabolism has diagnostic and therapeutic value for B cell malignancies with TRAF3 deletions or relevant mutations.
Collapse
Affiliation(s)
- Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ 08544.,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901; and
| | - Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854
| | - Yingying Liu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901; and.,W.M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ 08544.,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901; and
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854; .,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901; and
| |
Collapse
|
23
|
Kall SL, Whitlatch K, Smithgall TE, Lavie A. Molecular basis for the interaction between human choline kinase alpha and the SH3 domain of the c-Src tyrosine kinase. Sci Rep 2019; 9:17121. [PMID: 31745227 PMCID: PMC6864063 DOI: 10.1038/s41598-019-53447-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/30/2019] [Indexed: 01/09/2023] Open
Abstract
Choline kinase alpha is a 457-residue protein that catalyzes the reaction between ATP and choline to yield ADP and phosphocholine. This metabolic action has been well studied because of choline kinase's link to cancer malignancy and poor patient prognosis. As the myriad of x-ray crystal structures available for this enzyme show, chemotherapeutic drug design has centered on stopping the catalytic activity of choline kinase and reducing the downstream metabolites it produces. Furthermore, these crystal structures only reveal the catalytic domain of the protein, residues 80-457. However, recent studies provide evidence for a non-catalytic protein-binding role for choline kinase alpha. Here, we show that choline kinase alpha interacts with the SH3 domain of c-Src. Co-precipitation assays, surface plasmon resonance, and crystallographic analysis of a 1.5 Å structure demonstrate that this interaction is specific and is mediated by the poly-proline region found N-terminal to the catalytic domain of choline kinase. Taken together, these data offer strong evidence that choline kinase alpha has a heretofore underappreciated role in protein-protein interactions, which offers an exciting new way to approach drug development against this cancer-enhancing protein.
Collapse
Affiliation(s)
- Stefanie L Kall
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, 60607, USA
| | - Kindra Whitlatch
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15219, USA
| | - Thomas E Smithgall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15219, USA
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, 60607, USA.
- The Jesse Brown VA Medical Center, Chicago, Illinois, 60612, USA.
| |
Collapse
|
24
|
Zimmerman T, Lacal JC, Ibrahim SA. Choline Kinase Emerges as a Promising Drug Target in Gram-Positive Bacteria. Front Microbiol 2019; 6:2146. [PMID: 31681254 PMCID: PMC6813931 DOI: 10.3389/fmicb.2019.02146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/30/2019] [Indexed: 01/04/2023] Open
Abstract
Both nosocomial pathogens, such as Streptococcus pneumoniae and Haemophilus influenzae and food-borne pathogens, such as Bacillus cereus and Clostridium perfringens are known to be detrimental to human and animal health. The effectiveness of currently used treatments for these pathogens becomes limited as resistant strains emerge. Therefore, new methods for eliminating bacterial pathogens must be developed continuously. This includes establishing novel targets to which drug discovery efforts could be focused. A promising method for discovering new drug targets in prokaryotes is to take advantage of the information available regarding the enzymatic pathways that have been established as drug targets in eukaryotic systems and explore the analogous pathways found in bacterial systems. This is an efficient strategy because the same inhibitors developed at considerable expense to block these pathways in eukaryotic systems could also be employed in prokaryotes. Drugs that are used to prevent diseases involving eukaryotic cells could be repurposed as antibiotics and antimicrobials for the control of bacteria pathogens. This strategy could be pursued whenever the primary and tertiary structures of a target are are conserved between eukaryotic and prokaryotes. A possible novel target fitting these parameters is choline kinase (ChoK), whose active site sequences are conserved (Figure 1) and whose tertiary structure (Figure 2) is maintained. Here, we describe why ChoK is a putative drug target by describing its role in the growth and pathogenesis of Gram-positive bacteria S. pneumoniae and the Gram-negative bacteria H. influenzae. Using S. pneumoniae as a model, we also present promising preliminary information that repurposing of drugs known to inhibit the human isoform of ChoK (hChoK), is a promising strategy for blocking the growth of S. pneumoniae cells and inhibiting the activity of the S. pneumoniae isoform of ChoK (sChok), with downstream physiological effects on the cell wall.
Collapse
Affiliation(s)
- Tahl Zimmerman
- Food Microbiology and Biotechnology Laboratory, Food and Nutritional Sciences Program, North Carolina Agricultural and Technical State University, Greensboro, NC, United States
| | - Juan Carlos Lacal
- Department of Oncology, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Salam A Ibrahim
- Food Microbiology and Biotechnology Laboratory, Food and Nutritional Sciences Program, North Carolina Agricultural and Technical State University, Greensboro, NC, United States
| |
Collapse
|
25
|
Gougelet A, Sartor C, Senni N, Calderaro J, Fartoux L, Lequoy M, Wendum D, Talbot JN, Prignon A, Chalaye J, Imbeaud S, Zucman-Rossi J, Tordjmann T, Godard C, Bossard P, Rosmorduc O, Amaddeo G, Colnot S. Hepatocellular Carcinomas With Mutational Activation of Beta-Catenin Require Choline and Can Be Detected by Positron Emission Tomography. Gastroenterology 2019; 157:807-822. [PMID: 31194980 DOI: 10.1053/j.gastro.2019.05.069] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 05/18/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS In one-third of hepatocellular carcinomas (HCCs), cancer cells have mutations that activate β-catenin pathway. These cells have alterations in glutamine, bile, and lipid metabolism. We investigated whether positron emission tomography (PET) imaging allows identification of altered metabolic pathways that might be targeted therapeutically. METHODS We studied mice with activation of β-catenin in liver (Apcko-liv mice) and male C57Bl/6 mice given injections of diethylnitrosamine, which each develop HCCs. Mice were fed a conventional or a methionine- and choline-deficient diet or a choline-deficient (CD) diet. Choline uptake and metabolism in HCCs were analyzed by micro-PET imaging of mice; livers were collected and analyzed by histologic, metabolomic, messenger RNA quantification, and RNA-sequencing analyses. Fifty-two patients with HCC underwent PET imaging with 18F-fluorodeoxyglucose, followed by 18F-fluorocholine tracer metabolites. Human HCC specimens were analyzed by immunohistochemistry, quantitative polymerase chain reaction, and DNA sequencing. We used hepatocytes and mouse tumor explants for studies of incorporation of radiolabeled choline into phospholipids and its contribution to DNA methylation. We analyzed HCC progression in mice fed a CD diet. RESULTS Livers and tumors from Apcko-liv mice had increased uptake of dietary choline, which contributes to phospholipid formation and DNA methylation in hepatocytes. In patients and in mice, HCCs with activated β-catenin were positive in 18F-fluorocholine PET, but not 18F-fluorodeoxyglucose PET, and they overexpressed the choline transporter organic cation transporter 3. The HCC cells from Apcko-liv mice incorporated radiolabeled methyl groups of choline into phospholipids and DNA. In Apcko-liv mice, the methionine- and choline-deficient diet reduced proliferation and DNA hypermethylation of hepatocytes and HCC cells, and the CD diet reduced long-term progression of tumors. CONCLUSIONS In mice and humans, HCCs with mutations that activate β-catenin are characterized by increased uptake of a fluorocholine tracer, but not 18F-fluorodeoxyglucose, revealed by PET. The increased uptake of choline by HCCs promotes phospholipid formation, DNA hypermethylation, and hepatocyte proliferation. In mice, the CD diet reverses these effects and promotes regression of HCCs that overexpress β-catenin.
Collapse
Affiliation(s)
- Angélique Gougelet
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique, Unité Mixte De Recherché 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale Unité Mixte De Recherche 1138, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Chiara Sartor
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique, Unité Mixte De Recherché 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nadia Senni
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique, Unité Mixte De Recherché 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Julien Calderaro
- Assistance Publique-Hôpitaux de Paris, Département de Pathologie, Hôpital Universitaire Henri Mondor, Créteil, France; Institut National de la Santé et de la Recherche Médicale U955, Team 18, Institut Mondor de Recherche Biomédicale; Université Paris Est Créteil, Créteil, France
| | - Laetitia Fartoux
- Assistance Publique-Hôpitaux de Paris, Service d'Hépatologie, Hôpital St-Antoine, Sorbonne Université, Paris, France; Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche Saint-Antoine, Paris, France
| | - Marie Lequoy
- Assistance Publique-Hôpitaux de Paris, Service d'Hépatologie, Hôpital St-Antoine, Sorbonne Université, Paris, France; Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche Saint-Antoine, Paris, France
| | - Dominique Wendum
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche Saint-Antoine, Paris, France; Assistance Publique-Hôpitaux de Paris, Anatomie Pathologique, Hôpital St-Antoine, Sorbonne Université, Paris, France
| | - Jean-Noël Talbot
- Assistance Publique-Hôpitaux de Paris, Médecine Nucléaire, Hôpital Tenon, Sorbonne Université, Paris, France; Laboratoire d'Imagerie Moléculaire Photonique, UMS28, Phénotypage du Petit Animal, Sorbonne Université, Paris, France
| | - Aurélie Prignon
- Laboratoire d'Imagerie Moléculaire Photonique, UMS28, Phénotypage du Petit Animal, Sorbonne Université, Paris, France
| | - Julia Chalaye
- Assistance Publique-Hôpitaux de Paris, Médecine Nucléaire, Hôpital Universitaire Henri Mondor, Créteil, France
| | - Sandrine Imbeaud
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte De Recherché 1162, Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue Contre le Cancer, Institut Universitaire d'Hematologie, Paris, France
| | - Jessica Zucman-Rossi
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte De Recherché 1162, Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue Contre le Cancer, Institut Universitaire d'Hematologie, Paris, France
| | - Thierry Tordjmann
- Institut National de la Santé et de la Recherche Médicale U1174, Université Paris Sud, Orsay, France
| | - Cécile Godard
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique, Unité Mixte De Recherché 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale Unité Mixte De Recherche 1138, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Pascale Bossard
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique, Unité Mixte De Recherché 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale Unité Mixte De Recherche 1138, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Olivier Rosmorduc
- Assistance Publique-Hôpitaux de Paris, Service d'Hépatologie, Hôpital St-Antoine, Sorbonne Université, Paris, France; Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche Saint-Antoine, Paris, France
| | - Giuliana Amaddeo
- Assistance Publique-Hôpitaux de Paris, Médecine Interne, Hôpital Universitaire Henri Mondor, Créteil, France
| | - Sabine Colnot
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique, Unité Mixte De Recherché 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale Unité Mixte De Recherche 1138, Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
26
|
Sanchez-Lopez E, Zhong Z, Stubelius A, Sweeney SR, Booshehri LM, Antonucci L, Liu-Bryan R, Lodi A, Terkeltaub R, Lacal JC, Murphy AN, Hoffman HM, Tiziani S, Guma M, Karin M. Choline Uptake and Metabolism Modulate Macrophage IL-1β and IL-18 Production. Cell Metab 2019; 29:1350-1362.e7. [PMID: 30982734 PMCID: PMC6675591 DOI: 10.1016/j.cmet.2019.03.011] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/16/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023]
Abstract
Choline is a vitamin-like nutrient that is taken up via specific transporters and metabolized by choline kinase, which converts it to phosphocholine needed for de novo synthesis of phosphatidylcholine (PC), the main phospholipid of cellular membranes. We found that Toll-like receptor (TLR) activation enhances choline uptake by macrophages and microglia through induction of the choline transporter CTL1. Inhibition of CTL1 expression or choline phosphorylation attenuated NLRP3 inflammasome activation and IL-1β and IL-18 production in stimulated macrophages. Mechanistically, reduced choline uptake altered mitochondrial lipid profile, attenuated mitochondrial ATP synthesis, and activated the energy sensor AMP-activated protein kinase (AMPK). By potentiating mitochondrial recruitment of DRP1, AMPK stimulates mitophagy, which contributes to termination of NLRP3 inflammasome activation. Correspondingly, choline kinase inhibitors ameliorated acute and chronic models of IL-1β-dependent inflammation.
Collapse
Affiliation(s)
- Elsa Sanchez-Lopez
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, 92037, USA
| | - Zhenyu Zhong
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, 92037, USA; Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas TX 75390, USA
| | - Alexandra Stubelius
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, 92037, USA
| | - Shannon R Sweeney
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, 78723-3092, USA
| | - Laela M Booshehri
- Department of Pediatrics and Rady Children's Hospital, University of California San Diego, La Jolla, CA, 92037, USA
| | - Laura Antonucci
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, 92037, USA
| | - Ru Liu-Bryan
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, 92037, USA; VA San Diego Healthcare System, University of California San Diego, La Jolla, CA, 92037, USA
| | - Alessia Lodi
- Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, 78723-3092, USA; Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, 78723-3092, USA
| | - Robert Terkeltaub
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, 92037, USA; VA San Diego Healthcare System, University of California San Diego, La Jolla, CA, 92037, USA
| | - Juan Carlos Lacal
- Translational Oncology, Department of Oncology, Hospital Universitario Fuenlabrada, Instituto de Investigación Sanitaria IdiPAZ, Madrid, Spain
| | - Anne N Murphy
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, 92037, USA
| | - Hal M Hoffman
- Department of Pediatrics and Rady Children's Hospital, University of California San Diego, La Jolla, CA, 92037, USA
| | - Stefano Tiziani
- Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, 78723-3092, USA; Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, 78723-3092, USA; Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723-3092, USA
| | - Monica Guma
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, 92037, USA
| | - Michael Karin
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, 92037, USA.
| |
Collapse
|
27
|
Pillai SR, Damaghi M, Marunaka Y, Spugnini EP, Fais S, Gillies RJ. Causes, consequences, and therapy of tumors acidosis. Cancer Metastasis Rev 2019; 38:205-222. [PMID: 30911978 PMCID: PMC6625890 DOI: 10.1007/s10555-019-09792-7] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
While cancer is commonly described as "a disease of the genes," it is also associated with massive metabolic reprogramming that is now accepted as a disease "Hallmark." This programming is complex and often involves metabolic cooperativity between cancer cells and their surrounding stroma. Indeed, there is emerging clinical evidence that interrupting a cancer's metabolic program can improve patients' outcomes. The most commonly observed and well-studied metabolic adaptation in cancers is the fermentation of glucose to lactic acid, even in the presence of oxygen, also known as "aerobic glycolysis" or the "Warburg Effect." Much has been written about the mechanisms of the Warburg effect, and this remains a topic of great debate. However, herein, we will focus on an important sequela of this metabolic program: the acidification of the tumor microenvironment. Rather than being an epiphenomenon, it is now appreciated that this acidosis is a key player in cancer somatic evolution and progression to malignancy. Adaptation to acidosis induces and selects for malignant behaviors, such as increased invasion and metastasis, chemoresistance, and inhibition of immune surveillance. However, the metabolic reprogramming that occurs during adaptation to acidosis also introduces therapeutic vulnerabilities. Thus, tumor acidosis is a relevant therapeutic target, and we describe herein four approaches to accomplish this: (1) neutralizing acid directly with buffers, (2) targeting metabolic vulnerabilities revealed by acidosis, (3) developing acid-activatable drugs and nanomedicines, and (4) inhibiting metabolic processes responsible for generating acids in the first place.
Collapse
Affiliation(s)
- Smitha R Pillai
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33602, USA
| | - Mehdi Damaghi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33602, USA
| | - Yoshinori Marunaka
- Research Institute for Clinical Physiology, Kyoto, 604-8472, Japan
- Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, 525-8577, Japan
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | | | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), Viale Regina Elena, 299, 00161, Rome, Italy.
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33602, USA.
| |
Collapse
|
28
|
Abstract
Phospholipases D (PLDs) catalyze hydrolysis of the diester bond of phospholipids to generate phosphatidic acid and the free lipid headgroup. In mammals, PLD enzymes comprise the intracellular enzymes PLD1 and PLD2 and possibly the proteins encoded by related genes, as well as a class of cell surface and secreted enzymes with structural homology to ectonucleotide phosphatases/phosphodiesterases as typified by autotaxin (ENPP2) that have lysoPLD activities. Genetic and pharmacological loss-of-function approaches implicate these enzymes in intra- and intercellular signaling mediated by the lipid products phosphatidic acid, lysophosphatidic acid, and their metabolites, while the possibility that the water-soluble product of their reactions is biologically relevant has received far less attention. PLD1 and PLD2 are highly selective for phosphatidylcholine (PC), whereas autotaxin has broader substrate specificity for lysophospholipids but by virtue of the high abundance of lysophosphatidylcholine (LPC) in extracellular fluids predominantly hydrolyses this substrate. In all cases, the water-soluble product of these PLD activities is choline. Although choline can be formed de novo by methylation of phosphatidylethanolamine, this activity is absent in most tissues, so mammals are effectively auxotrophic for choline. Dietary consumption of choline in both free and esterified forms is substantial. Choline is necessary for synthesis of the neurotransmitter acetylcholine and of the choline-containing phospholipids PC and sphingomyelin (SM) and also plays a recently appreciated important role as a methyl donor in the pathways of "one-carbon (1C)" metabolism. This review discusses emerging evidence that some of the biological functions of these intra- and extracellular PLD enzymes involve generation of choline with a particular focus on the possibility that these choline and PLD dependent processes are dysregulated in cancer.
Collapse
|
29
|
Choline Kinase Alpha Inhibition by EB-3D Triggers Cellular Senescence, Reduces Tumor Growth and Metastatic Dissemination in Breast Cancer. Cancers (Basel) 2018; 10:cancers10100391. [PMID: 30360374 PMCID: PMC6209942 DOI: 10.3390/cancers10100391] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 12/15/2022] Open
Abstract
Choline kinase (ChoK) is the first enzyme of the Kennedy pathway leading to the biosynthesis of phosphatidylcholine (PtdCho), the most abundant phospholipid in eukaryotic cell membranes. EB-3D is a novel choline kinase α1 (ChoKα1) inhibitor with potent antiproliferative activity against a panel of several cancer cell lines. ChoKα1 is particularly overexpressed and hyperactivated in aggressive breast cancer. By NMR analysis, we demonstrated that EB-3D is able to reduce the synthesis of phosphocholine, and using flow cytometry, immunoblotting, and q-RT-PCR as well as proliferation and invasion assays, we proved that EB-3D strongly impairs breast cancer cell proliferation, migration, and invasion. EB-3D induces senescence in breast cancer cell lines through the activation of the metabolic sensor AMPK and the subsequent dephosphorylation of mTORC1 downstream targets, such as p70S6K, S6 ribosomal protein, and 4E-BP1. Moreover, EB-3D strongly synergizes with drugs commonly used for breast cancer treatment. The antitumorigenic potential of EB-3D was evaluated in vivo in the syngeneic orthotopic E0771 mouse model of breast cancer, where it induces a significant reduction of the tumor mass at low doses. In addition, EB-3D showed an antimetastatic effect in experimental and spontaneous metastasis models. Altogether, our results indicate that EB-3D could be a promising new anticancer agent to improve aggressive breast cancer treatment protocols.
Collapse
|
30
|
Lead optimization-hit expansion of new asymmetrical pyridinium/quinolinium compounds as choline kinase α1 inhibitors. Future Med Chem 2018; 10:1769-1786. [PMID: 30043647 DOI: 10.4155/fmc-2018-0059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM Choline kinase α inhibitors represent one of the newest classes of cytotoxic drugs for cancer treatment, since aberrant choline metabolism is a characteristic shared by many human cancers. RESULTS Here, we present a new class of asymmetrical pyridinium/quinolinium derivatives developed and designed based on drug optimization. CONCLUSION Among all compounds described here, compound 8, bearing a 7-chloro-4N-methyl-p-chloroaniline quinolinium moiety, exhibited the greatest inhibitory activity at the enzyme (IC50 = 0.29 μM) and antiproliferative activity in cellular assays (GI50 = 0.29-0.92 μM). Specifically, compound 8 strongly induces a cell-cycle arrest in G1 phase, but it does not significantly induce apoptosis while causing senescence in the MDA-MB-231 cell line.
Collapse
|
31
|
One-Carbon Metabolism: Biological Players in Epithelial Ovarian Cancer. Int J Mol Sci 2018; 19:ijms19072092. [PMID: 30029471 PMCID: PMC6073728 DOI: 10.3390/ijms19072092] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/06/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023] Open
Abstract
Metabolism is deeply involved in cell behavior and homeostasis maintenance, with metabolites acting as molecular intermediates to modulate cellular functions. In particular, one-carbon metabolism is a key biochemical pathway necessary to provide carbon units required for critical processes, including nucleotide biosynthesis, epigenetic methylation, and cell redox-status regulation. It is, therefore, not surprising that alterations in this pathway may acquire fundamental importance in cancer onset and progression. Two of the major actors in one-carbon metabolism, folate and choline, play a key role in the pathobiology of epithelial ovarian cancer (EOC), the deadliest gynecological malignancy. EOC is characterized by a cholinic phenotype sustained via increased activity of choline kinase alpha, and via membrane overexpression of the alpha isoform of the folate receptor (FRα), both of which are known to contribute to generating regulatory signals that support EOC cell aggressiveness and proliferation. Here, we describe in detail the main biological processes associated with one-carbon metabolism, and the current knowledge about its role in EOC. Moreover, since the cholinic phenotype and FRα overexpression are unique properties of tumor cells, but not of normal cells, they can be considered attractive targets for the development of therapeutic approaches.
Collapse
|
32
|
Hu L, Wang RY, Cai J, Feng D, Yang GZ, Xu QG, Zhai YX, Zhang Y, Zhou WP, Cai QP. Overexpression of CHKA contributes to tumor progression and metastasis and predicts poor prognosis in colorectal carcinoma. Oncotarget 2018; 7:66660-66678. [PMID: 27556502 PMCID: PMC5341828 DOI: 10.18632/oncotarget.11433] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/13/2016] [Indexed: 12/13/2022] Open
Abstract
Aberrant expression of choline kinase alpha (CHKA) has been reported in a variety of human malignancies including colorectal carcinoma (CRC). However, the role of CHKA in the progression and prognosis of CRC remains unknown. In this study, we found that CHKA was frequently upregulated in CRC clinical samples and CRC-derived cell lines and was significantly correlated with lymph node metastasis (p = 0.028), TNM stage (p = 0.009), disease recurrence (p = 0.004) and death (p < 0.001). Survival analyses indicated that patients with higher CHKA expression had a significantly shorter disease-free survival (DFS) and disease-specific survival (DSS) than those with lower CHKA expression. Multivariate analyses confirmed that increased CHKA expression was an independent unfavorable prognostic factor for CRC patients. In addition, combination of CHKA with TNM stage was a more powerful predictor of poor prognosis than either parameter alone. Functional study demonstrated that knockdown of CHKA expression profoundly suppressed the growth and metastasis of CRC cells both in vitro and in vivo. Mechanistic investigation revealed that EGFR/PI3K/AKT pathway was essential for mediating CHKA function. In conclusion, our results provide the first evidence that CHKA contributes to tumor progression and metastasis and may serve as a novel prognostic biomarker and potential therapeutic target in CRC.
Collapse
Affiliation(s)
- Liang Hu
- Anal-Colorectal Surgery Institute, 150th Hospital of PLA, Luoyang, China.,Department of Gastrointestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ruo-Yu Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jian Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan Feng
- Department of Oncology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Guang-Zhen Yang
- Department of Clinical Laboratory, 150th Hospital of PLA, Luoyang, China
| | - Qing-Guo Xu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yan-Xia Zhai
- Anal-Colorectal Surgery Institute, 150th Hospital of PLA, Luoyang, China
| | - Yu Zhang
- Anal-Colorectal Surgery Institute, 150th Hospital of PLA, Luoyang, China
| | - Wei-Ping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Qing-Ping Cai
- Department of Gastrointestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
33
|
Kall SL, Delikatny EJ, Lavie A. Identification of a Unique Inhibitor-Binding Site on Choline Kinase α. Biochemistry 2018; 57:1316-1325. [PMID: 29389115 DOI: 10.1021/acs.biochem.7b01257] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Choline kinase α (ChoKα) is an enzyme that is upregulated in many types of cancer and has been shown to be tumorigenic. As such, it makes a promising target for inhibiting tumor growth. Though there have been several inhibitors synthesized for ChoKα, not all of them demonstrate the same efficacy in vivo, though the reasons behind this difference in potency are not clear. One particular inhibitor, designated TCD-717, has recently completed phase I clinical trials. Cell culture and in vitro studies support the powerful inhibitory effect TCD-717 has on ChoKα, but an examination of the inhibitor's interaction with the ChoKα enzyme has been missing prior to this work. Here we detail the 2.35 Å structure of ChoKα in complex with TCD-717. Examination of this structure in conjunction with kinetic assays reveals that TCD-717 does not bind directly in the choline pocket as do previously characterized ChoKα inhibitors, but rather in a proximal but novel location near the surface of the enzyme. The unique binding site identified for TCD-717 lends insight for the future design of more potent in vivo inhibitors for ChoKα.
Collapse
Affiliation(s)
- Stefanie L Kall
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, Illinois 60607, United States
| | - Edward J Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania 19083, United States
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, Illinois 60607, United States.,The Jesse Brown VA Medical Center , Chicago, Illinois 60612, United States
| |
Collapse
|
34
|
Arlauckas SP, Kumar M, Popov AV, Poptani H, Delikatny EJ. Near infrared fluorescent imaging of choline kinase alpha expression and inhibition in breast tumors. Oncotarget 2017; 8:16518-16530. [PMID: 28157707 PMCID: PMC5369982 DOI: 10.18632/oncotarget.14965] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
Choline kinase alpha (ChoKα) overexpression is associated with an aggressive tumor phenotype. ChoKα inhibitors induce apoptosis in tumors, however validation of their specificity is difficult in vivo. We report the use of optical imaging to assess ChoKα status in cells and in vivo using JAS239, a carbocyanine-based ChoKα inhibitor with inherent near infrared fluorescence. JAS239 attenuated choline phosphorylation and viability in a panel of human breast cancer cell lines. Antibody blockade prevented cellular retention of JAS239 indicating direct interaction with ChoKα independent of the choline transporters and catabolic choline pathways. In mice bearing orthotopic MCF7 breast xenografts, optical imaging with JAS239 distinguished tumors overexpressing ChoKα from their empty vector counterparts and delineated tumor margins. Pharmacological inhibition of ChoK by the established inhibitor MN58b led to a growth inhibition in 4175-Luc+ tumors that was accompanied by concomitant reduction in JAS239 uptake and decreased total choline metabolite levels as measured using magnetic resonance spectroscopy. At higher therapeutic doses, JAS239 was as effective as MN58b at arresting tumor growth and inducing apoptosis in MDA-MB-231 tumors, significantly reducing tumor choline below baseline levels without observable systemic toxicity. These data introduce a new method to monitor therapeutically effective inhibitors of choline metabolism in breast cancer using a small molecule companion diagnostic.
Collapse
Affiliation(s)
- Sean P Arlauckas
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Manoj Kumar
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anatoliy V Popov
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Harish Poptani
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Cellular and Molecular Physiology, Institute of Regenerative Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Edward J Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
35
|
Metabolic Portraits of Breast Cancer by HR MAS MR Spectroscopy of Intact Tissue Samples. Metabolites 2017; 7:metabo7020018. [PMID: 28509845 PMCID: PMC5487989 DOI: 10.3390/metabo7020018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 04/20/2017] [Accepted: 05/09/2017] [Indexed: 12/17/2022] Open
Abstract
Despite progress in early detection and therapeutic strategies, breast cancer remains the second leading cause of cancer-related death among women globally. Due to the heterogeneity and complexity of tumor biology, breast cancer patients with similar diagnosis might have different prognosis and response to treatment. Thus, deeper understanding of individual tumor properties is necessary. Cancer cells must be able to convert nutrients to biomass while maintaining energy production, which requires reprogramming of central metabolic processes in the cells. This phenomenon is increasingly recognized as a potential target for treatment, but also as a source for biomarkers that can be used for prognosis, risk stratification and therapy monitoring. Magnetic resonance (MR) metabolomics is a widely used approach in translational research, aiming to identify clinically relevant metabolic biomarkers or generate novel understanding of the molecular biology in tumors. Ex vivo proton high-resolution magic angle spinning (HR MAS) MR spectroscopy is widely used to study central metabolic processes in a non-destructive manner. Here we review the current status for HR MAS MR spectroscopy findings in breast cancer in relation to glucose, amino acid and choline metabolism.
Collapse
|
36
|
Cheng M, Bhujwalla ZM, Glunde K. Targeting Phospholipid Metabolism in Cancer. Front Oncol 2016; 6:266. [PMID: 28083512 PMCID: PMC5187387 DOI: 10.3389/fonc.2016.00266] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022] Open
Abstract
All cancers tested so far display abnormal choline and ethanolamine phospholipid metabolism, which has been detected with numerous magnetic resonance spectroscopy (MRS) approaches in cells, animal models of cancer, as well as the tumors of cancer patients. Since the discovery of this metabolic hallmark of cancer, many studies have been performed to elucidate the molecular origins of deregulated choline metabolism, to identify targets for cancer treatment, and to develop MRS approaches that detect choline and ethanolamine compounds for clinical use in diagnosis and treatment monitoring. Several enzymes in choline, and recently also ethanolamine, phospholipid metabolism have been identified, and their evaluation has shown that they are involved in carcinogenesis and tumor progression. Several already established enzymes as well as a number of emerging enzymes in phospholipid metabolism can be used as treatment targets for anticancer therapy, either alone or in combination with other chemotherapeutic approaches. This review summarizes the current knowledge of established and relatively novel targets in phospholipid metabolism of cancer, covering choline kinase α, phosphatidylcholine-specific phospholipase D1, phosphatidylcholine-specific phospholipase C, sphingomyelinases, choline transporters, glycerophosphodiesterases, phosphatidylethanolamine N-methyltransferase, and ethanolamine kinase. These enzymes are discussed in terms of their roles in oncogenic transformation, tumor progression, and crucial cancer cell properties such as fast proliferation, migration, and invasion. Their potential as treatment targets are evaluated based on the current literature.
Collapse
Affiliation(s)
- Menglin Cheng
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Glunde
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Human Choline Kinase-α Promotes Hepatitis C Virus RNA Replication through Modulation of Membranous Viral Replication Complex Formation. J Virol 2016; 90:9075-95. [PMID: 27489281 PMCID: PMC5044849 DOI: 10.1128/jvi.00960-16] [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: 05/17/2016] [Accepted: 07/20/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Hepatitis C virus (HCV) infection reorganizes cellular membranes to create an active viral replication site named the membranous web (MW). The role that human choline kinase-α (hCKα) plays in HCV replication remains elusive. Here, we first showed that hCKα activity, not the CDP-choline pathway, promoted viral RNA replication. Confocal microscopy and subcellular fractionation of HCV-infected cells revealed that a small fraction of hCKα colocalized with the viral replication complex (RC) on the endoplasmic reticulum (ER) and that HCV infection increased hCKα localization to the ER. In the pTM-NS3-NS5B model, NS3-NS5B expression increased the localization of the wild-type, not the inactive D288A mutant, hCKα on the ER, and hCKα activity was required for effective trafficking of hCKα and NS5A to the ER. Coimmunoprecipitation showed that hCKα was recruited onto the viral RC presumably through its binding to NS5A domain 1 (D1). hCKα silencing or treatment with CK37, an hCKα activity inhibitor, abolished HCV-induced MW formation. In addition, hCKα depletion hindered NS5A localization on the ER, interfered with NS5A and NS5B colocalization, and mitigated NS5A-NS5B interactions but had no apparent effect on NS5A-NS4B and NS4B-NS5B interactions. Nevertheless, hCKα activity was not essential for the binding of NS5A to hCKα or NS5B. These findings demonstrate that hCKα forms a complex with NS5A and that hCKα activity enhances the targeting of the complex to the ER, where hCKα protein, not activity, mediates NS5A binding to NS5B, thereby promoting functional membranous viral RC assembly and viral RNA replication. IMPORTANCE HCV infection reorganizes the cellular membrane to create an active viral replication site named the membranous web (MW). Here, we report that human choline kinase-α (hCKα) acts as an essential host factor for HCV RNA replication. A fraction of hCKα colocalizes with the viral replication complex (RC) on the endoplasmic reticulum (ER) in HCV-infected cells. NS3-NS5B expression increases ER localization of wild-type, but not D288A mutant, hCKα, and hCKα activity facilitates the transport of itself and NS5A to the ER. Silencing or inactivation of hCKα abrogates MW formation. Moreover, hCKα is recruited by NS5A independent of hCKα activity, presumably through binding to NS5A D1. hCKα activity then mediates the ER targeting of the hCKα-NS5A complex. On the ER membrane, hCKα protein, per se, induces NS5A binding to NS5B, thereby promoting membranous RC formation and viral RNA replication. Our study may benefit the development of hCKα-targeted anti-HCV therapeutics.
Collapse
|
38
|
Zhang L, Chen P, Yang S, Li G, Bao W, Wu P, Jiang S. CHKA mediates the poor prognosis of lung adenocarcinoma and acts as a prognostic indicator. Oncol Lett 2016; 12:1849-1853. [PMID: 27588131 PMCID: PMC4998128 DOI: 10.3892/ol.2016.4810] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/10/2016] [Indexed: 01/04/2023] Open
Abstract
Choline kinase α (CHKA), the enzyme that converts choline to phosphocholine, has been studied in human carcinogenesis widely. However, the expression and underlying clinicopathological characteristics of CHKA in lung adenocarcinoma remains elusive. In the present study, a tissue microarray of 119 pairs of lung adenocarcinoma samples and corresponding adjacent normal mucosae was used to analysis CHKA expression by immunohistochemistry, and CHKA was observed to exhibit enhanced expression in lung adenocarcinoma tissues. Elevated CHKA expression in lung adenocarcinoma tissues at the gene and protein level was observed. The levels of CHKA expression were closely associated with the poor prognosis status of lung adenocarcinoma patients. Furthermore, certain clinicopathological characteristics such as tumor diameter and differentiation were observed to be significant in those lung adenocarcinoma patients who displayed enhanced CHKA expression. The analysis of CHKA expression could provide a more precise way to predict the prognosis of lung adenocarcinoma patients. Collectively, the present study revealed a novel biomarker in lung adenocarcinoma, and indicated that CHKA may be a promising prognostic marker and therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Li Zhang
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China; Department of Respiratory Intensive Care Unit, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Ping Chen
- Department of Gastroenterology, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Shen Yang
- Department of Neurology, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Guodong Li
- Department of Respiratory Medicine, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Wentao Bao
- Department of Respiratory Intensive Care Unit, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Peng Wu
- Department of Respiratory Intensive Care Unit, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Shujuan Jiang
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
39
|
Arlauckas SP, Popov AV, Delikatny EJ. Choline kinase alpha-Putting the ChoK-hold on tumor metabolism. Prog Lipid Res 2016; 63:28-40. [PMID: 27073147 PMCID: PMC5360181 DOI: 10.1016/j.plipres.2016.03.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/14/2016] [Accepted: 03/26/2016] [Indexed: 12/24/2022]
Abstract
It is well established that lipid metabolism is drastically altered during tumor development and response to therapy. Choline kinase alpha (ChoKα) is a key mediator of these changes, as it represents the first committed step in the Kennedy pathway of phosphatidylcholine biosynthesis and ChoKα expression is upregulated in many human cancers. ChoKα activity is associated with drug resistant, metastatic, and malignant phenotypes, and represents a robust biomarker and therapeutic target in cancer. Effective ChoKα inhibitors have been developed and have recently entered clinical trials. ChoKα's clinical relevance was, until recently, attributed solely to its production of second messenger intermediates of phospholipid synthesis. The recent discovery of a non-catalytic scaffolding function of ChoKα may link growth receptor signaling to lipid biogenesis and requires a reinterpretation of the design and validation of ChoKα inhibitors. Advances in positron emission tomography, magnetic resonance spectroscopy, and optical imaging methods now allow for a comprehensive understanding of ChoKα expression and activity in vivo. We will review the current understanding of ChoKα metabolism, its role in tumor biology and the development and validation of targeted therapies and companion diagnostics for this important regulatory enzyme. This comes at a critical time as ChoKα-targeting programs receive more clinical interest.
Collapse
Affiliation(s)
- Sean P Arlauckas
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anatoliy V Popov
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E James Delikatny
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
40
|
Bagnoli M, Granata A, Nicoletti R, Krishnamachary B, Bhujwalla ZM, Canese R, Podo F, Canevari S, Iorio E, Mezzanzanica D. Choline Metabolism Alteration: A Focus on Ovarian Cancer. Front Oncol 2016; 6:153. [PMID: 27446799 PMCID: PMC4916225 DOI: 10.3389/fonc.2016.00153] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/07/2016] [Indexed: 12/31/2022] Open
Abstract
Compared with normal differentiated cells, cancer cells require a metabolic reprograming to support their high proliferation rates and survival. Aberrant choline metabolism is a fairly new metabolic hallmark reflecting the complex reciprocal interactions between oncogenic signaling and cellular metabolism. Alterations of the involved metabolic network may be sustained by changes in activity of several choline transporters as well as of enzymes such as choline kinase-alpha (ChoK-α) and phosphatidylcholine-specific phospholipases C and D. Of note, the net outcome of these enzymatic alterations is an increase of phosphocholine and total choline-containing compounds, a "cholinic phenotype" that can be monitored in cancer by magnetic resonance spectroscopy. This review will highlight the molecular basis for targeting this pathway in epithelial ovarian cancer (EOC), a highly heterogeneous and lethal malignancy characterized by late diagnosis, frequent relapse, and development of chemoresistance. Modulation of ChoK-α expression impairs only EOC but not normal ovarian cells, thus supporting the hypothesis that "cholinic phenotype" is a peculiar feature of transformed cells and indicating ChoK-α targeting as a novel approach to improve efficacy of standard EOC chemotherapeutic treatments.
Collapse
Affiliation(s)
- Marina Bagnoli
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| | - Anna Granata
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| | - Roberta Nicoletti
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| | - Balaji Krishnamachary
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, In Vivo Cellular and Molecular Imaging Center, The Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Zaver M Bhujwalla
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, In Vivo Cellular and Molecular Imaging Center, The Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Rossella Canese
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Franca Podo
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Silvana Canevari
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Functional Genomics and Informatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Egidio Iorio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Delia Mezzanzanica
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| |
Collapse
|
41
|
Design, synthesis, crystallization and biological evaluation of new symmetrical biscationic compounds as selective inhibitors of human Choline Kinase α1 (ChoKα1). Sci Rep 2016; 6:23793. [PMID: 27029499 PMCID: PMC4814829 DOI: 10.1038/srep23793] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/14/2016] [Indexed: 01/20/2023] Open
Abstract
A novel family of compounds derivative of 1,1′-(((ethane-1,2-diylbis(oxy))bis(4,1-phenylene))bis(methylene))-bispyridinium or –bisquinolinium bromide (10a-l) containing a pair of oxygen atoms in the spacer of the linker between the biscationic moieties, were synthesized and evaluated as inhibitors of choline kinase against a panel of cancer-cell lines. The most promising compounds in this series were 1,1′-(((ethane-1,2-diylbis(oxy))bis(4,1-phenylene))bis(methylene))bis(4-(dimethylamino)pyridinium) bromide (10a) and 1,1′-(((ethane-1,2-diylbis(oxy))bis(4,1-phenylene))bis(methylene))-bis(7-chloro-4-(pyrrolidin-1-yl)quinolinium) bromide (10l), which inhibit human choline kinase (ChoKα1) with IC50 of 1.0 and 0.92 μM, respectively, in a range similar to that of the previously reported biscationic compounds MN58b and RSM932A. Our compounds show greater antiproliferative activities than do the reference compounds, with unprecedented values of GI50 in the nanomolar range for several of the cancer-cell lines assayed, and more importantly they present low toxicity in non-tumoral cell lines, suggesting a cancer-cell-selective antiproliferative activity. Docking studies predict that the compounds interact with the choline-binding site in agreement with the binding mode of most previously reported biscationic compounds. Moreover, the crystal structure of ChoKα1 with compound 10a reveals that this compound binds to the choline-binding site and mimics HC-3 binding mode as never before.
Collapse
|
42
|
Smith TAD, Phyu SM. Metformin Decouples Phospholipid Metabolism in Breast Cancer Cells. PLoS One 2016; 11:e0151179. [PMID: 26959405 PMCID: PMC4784930 DOI: 10.1371/journal.pone.0151179] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/24/2016] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION The antidiabetic drug metformin, currently undergoing trials for cancer treatment, modulates lipid and glucose metabolism both crucial in phospholipid synthesis. Here the effect of treatment of breast tumour cells with metformin on phosphatidylcholine (PtdCho) metabolism which plays a key role in membrane synthesis and intracellular signalling has been examined. METHODS MDA-MB-468, BT474 and SKBr3 breast cancer cell lines were treated with metformin and [3H-methyl]choline and [14C(U)]glucose incorporation and lipid accumulation determined in the presence and absence of lipase inhibitors. Activities of choline kinase (CK), CTP:phosphocholine cytidylyl transferase (CCT) and PtdCho-phospholipase C (PLC) were also measured. [3H] Radiolabelled metabolites were determined using thin layer chromatography. RESULTS Metformin-treated cells exhibited decreased formation of [3H]phosphocholine but increased accumulation of [3H]choline by PtdCho. CK and PLC activities were decreased and CCT activity increased by metformin-treatment. [14C] incorporation into fatty acids was decreased and into glycerol was increased in breast cancer cells treated with metformin incubated with [14C(U)]glucose. CONCLUSION This is the first study to show that treatment of breast cancer cells with metformin induces profound changes in phospholipid metabolism.
Collapse
Affiliation(s)
- Tim A. D. Smith
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Su M. Phyu
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| |
Collapse
|
43
|
Zech SG, Kohlmann A, Zhou T, Li F, Squillace RM, Parillon LE, Greenfield MT, Miller DP, Qi J, Thomas RM, Wang Y, Xu Y, Miret JJ, Shakespeare WC, Zhu X, Dalgarno DC. Novel Small Molecule Inhibitors of Choline Kinase Identified by Fragment-Based Drug Discovery. J Med Chem 2016; 59:671-86. [PMID: 26700752 DOI: 10.1021/acs.jmedchem.5b01552] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Choline kinase α (ChoKα) is an enzyme involved in the synthesis of phospholipids and thereby plays key roles in regulation of cell proliferation, oncogenic transformation, and human carcinogenesis. Since several inhibitors of ChoKα display antiproliferative activity in both cellular and animal models, this novel oncogene has recently gained interest as a promising small molecule target for cancer therapy. Here we summarize our efforts to further validate ChoKα as an oncogenic target and explore the activity of novel small molecule inhibitors of ChoKα. Starting from weakly binding fragments, we describe a structure based lead discovery approach, which resulted in novel highly potent inhibitors of ChoKα. In cancer cell lines, our lead compounds exhibit a dose-dependent decrease of phosphocholine, inhibition of cell growth, and induction of apoptosis at low micromolar concentrations. The druglike lead series presented here is optimizable for improvements in cellular potency, drug target residence time, and pharmacokinetic parameters. These inhibitors may be utilized not only to further validate ChoKα as antioncogenic target but also as novel chemical matter that may lead to antitumor agents that specifically interfere with cancer cell metabolism.
Collapse
Affiliation(s)
- Stephan G Zech
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Anna Kohlmann
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Tianjun Zhou
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Feng Li
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Rachel M Squillace
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Lois E Parillon
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Matthew T Greenfield
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - David P Miller
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Jiwei Qi
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - R Mathew Thomas
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Yihan Wang
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Yongjin Xu
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Juan J Miret
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - William C Shakespeare
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Xiaotian Zhu
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - David C Dalgarno
- ARIAD Pharmaceuticals, Inc. , 26 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
44
|
Mignion L, Danhier P, Magat J, Porporato PE, Masquelier J, Gregoire V, Muccioli GG, Sonveaux P, Gallez B, Jordan BF. Non-invasive in vivo imaging of early metabolic tumor response to therapies targeting choline metabolism. Int J Cancer 2015; 138:2043-9. [PMID: 26595604 DOI: 10.1002/ijc.29932] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 01/17/2023]
Abstract
The cholinic phenotype, characterized by elevated phosphocholine and a high production of total-choline (tCho)-containing metabolites, is a metabolic hallmark of cancer. It can be exploited for targeted therapy. Non-invasive imaging biomarkers are required to evaluate an individual's response to targeted anticancer agents that usually do not rapidly cause tumor shrinkage. Because metabolic changes can manifest at earlier stages of therapy than changes in tumor size, the aim of the current study was to evaluate (1)H-MRS and diffusion-weighted MRI (DW-MRI) as markers of tumor response to the modulation of the choline pathway in mammary tumor xenografts. Inhibition of choline kinase activity was achieved with the direct pharmacological inhibitor H-89, indirect inhibitor sorafenib and down-regulation of choline-kinase α (ChKA) expression using specific short-hairpin RNA (shRNA). While all three strategies significantly decreased tCho tumor content in vivo, only sorafenib and anti-ChKA shRNA significantly repressed tumor growth. The increase of apparent-diffusion-coefficient of water (ADCw) measured by DW-MRI, was predictive of the induced necrosis and inhibition of the tumor growth in sorafenib treated mice, while the absence of change in ADC values in H89 treated mice predicted the absence of effect in terms of tumor necrosis and tumor growth. In conclusion, (1)H-choline spectroscopy can be useful as a pharmacodynamic biomarker for choline targeted agents, while DW-MRI can be used as an early marker of effective tumor response to choline targeted therapies. DW-MRI combined to choline spectroscopy may provide a useful non-invasive marker for the early clinical assessment of tumor response to therapies targeting choline signaling.
Collapse
Affiliation(s)
- Lionel Mignion
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| | - Pierre Danhier
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| | - Julie Magat
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| | - Paolo E Porporato
- Pole of Pharmacology, Institut De Recherche Expérimentale Et Clinique (IREC), Université Catholique De Louvain (UCL), Avenue Mounier, 52 Box B1.53.09, Brussels, Belgium
| | - Julien Masquelier
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 72 Box B1.72.01, Brussels, Belgium
| | - Vincent Gregoire
- Center for Molecular Imaging, Radiotherapy and Oncology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Hippocrate, 55 Box B1.55.02, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 72 Box B1.72.01, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology, Institut De Recherche Expérimentale Et Clinique (IREC), Université Catholique De Louvain (UCL), Avenue Mounier, 52 Box B1.53.09, Brussels, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| |
Collapse
|
45
|
Estévez-Braun A, Ravelo AG, Pérez-Sacau E, Lacal JC. A new family of choline kinase inhibitors with antiproliferative and antitumor activity derived from natural products. Clin Transl Oncol 2014; 17:74-84. [DOI: 10.1007/s12094-014-1260-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/21/2014] [Indexed: 11/29/2022]
|
46
|
Pun WK, Chow SP, Fang D, Cheng CL, Leong JC, Ng C. Post-traumatic oedema of the foot after tibial fracture. Expert Rev Mol Diagn 1990; 15:735-47. [PMID: 2592102 DOI: 10.1586/14737159.2015.1039515] [Citation(s) in RCA: 103] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A total of 97 patients with diaphyseal tibial fractures treated with functional bracing were studied prospectively. Persistent ipsilateral foot swelling was present in 84.5 per cent of the patients. Most of the swellings subsided with time, but a small percentage of them persisted for a duration of 2 years or more after injury. The time for disappearance of the swelling in 50 per cent of the patients was 18.6 weeks. The development of oedema is not related to the age and sex of the patients, the configuration, type and level of the fractures, or the association of a fibular fracture. The bone healed quicker in those who did not have swelling of the foot. Once the swelling has developed, it seems to run its own course and its disappearance is not related to the age and sex, the configuration, type and level of fractures, the association of a fibular fracture, or the time for fracture healing. This complication does not have any adverse effect on the functional recovery of the patients.
Collapse
Affiliation(s)
- W K Pun
- Department of Orthopaedic Surgery, University of Hong Kong, Queen Mary Hospital
| | | | | | | | | | | |
Collapse
|