1
|
Jacquemin C, El Orch W, Diaz O, Lalande A, Aublin-Gex A, Jacolin F, Toesca J, Si-Tahar M, Mathieu C, Lotteau V, Perrin-Cocon L, Vidalain PO. Pharmacological induction of the hypoxia response pathway in Huh7 hepatoma cells limits proliferation but increases resilience under metabolic stress. Cell Mol Life Sci 2024; 81:320. [PMID: 39078527 PMCID: PMC11335246 DOI: 10.1007/s00018-024-05361-6] [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: 04/02/2024] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 07/31/2024]
Abstract
The hypoxia response pathway enables adaptation to oxygen deprivation. It is mediated by hypoxia-inducible factors (HIF), which promote metabolic reprogramming, erythropoiesis, angiogenesis and tissue remodeling. This led to the successful development of HIF-inducing drugs for treating anemia and some of these molecules are now in clinic. However, elevated levels of HIFs are frequently associated with tumor growth, poor prognosis, and drug resistance in various cancers, including hepatocellular carcinoma (HCC). Consequently, there are concerns regarding the recommendation of HIF-inducing drugs in certain clinical situations. Here, we analyzed the effects of two HIF-inducing drugs, Molidustat and Roxadustat, in the well-characterized HCC cell line Huh7. These drugs increased HIF-1α and HIF-2α protein levels which both participate in inducing hypoxia response genes such as BNIP3, SERPINE1, LDHA or EPO. Combined transcriptomics, proteomics and metabolomics showed that Molidustat increased the expression of glycolytic enzymes, while the mitochondrial network was fragmented and cellular respiration decreased. This metabolic remodeling was associated with a reduced proliferation and a lower demand for pyrimidine supply, but an increased ability of cells to convert pyruvate to lactate. This was accompanied by a higher resistance to the inhibition of mitochondrial respiration by antimycin A, a phenotype confirmed in Roxadustat-treated Huh7 cells and Molidustat-treated hepatoblastoma cells (Huh6 and HepG2). Overall, this study shows that HIF-inducing drugs increase the metabolic resilience of liver cancer cells to metabolic stressors, arguing for careful monitoring of patients treated with HIF-inducing drugs, especially when they are at risk of liver cancer.
Collapse
Affiliation(s)
- Clémence Jacquemin
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Walid El Orch
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Olivier Diaz
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Alexandre Lalande
- CIRI, Centre International de Recherche en Infectiologie, Team NeuroInvasion, Tropism and Viral Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Anne Aublin-Gex
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Florentine Jacolin
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Johan Toesca
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Mustapha Si-Tahar
- Centre d'Etude des Pathologies Respiratoires (CEPR), Faculty of Medecine, Inserm, U1100, 37000, Tours, France
| | - Cyrille Mathieu
- CIRI, Centre International de Recherche en Infectiologie, Team NeuroInvasion, Tropism and Viral Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Vincent Lotteau
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
- Laboratoire P4 INSERM-Jean Mérieux, Lyon, France
| | - Laure Perrin-Cocon
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France.
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France.
| |
Collapse
|
2
|
Banka VK, Sainas S, Martino E, Wang J, Lolli ML, Ding YS. Radiosynthesis of [ 18F]brequinar for in vivo PET imaging of hDHODH for potential studies of acute myeloid leukemia and cancers. RSC Med Chem 2024:d4md00433g. [PMID: 39149561 PMCID: PMC11320022 DOI: 10.1039/d4md00433g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/21/2024] [Indexed: 08/17/2024] Open
Abstract
Dihydroorotate dehydrogenase (DHODH), an enzyme that plays a critical role in the de novo pyrimidine biosynthesis, has been recognized as a promising target for the treatment of diseases that involve cellular proliferation, such as autoimmune diseases and cancers. Pharmacological inhibition of human DHODH (hDHODH) that offers a potential therapeutic strategy for the treatment in adult subjects with acute myeloid leukemia (AML) has recently been supported by phase I/II clinical trials for the treatment of patients with relapsed/refractory AML. To facilitate the development of optimized hDHODH inhibitors, the presence of an in vivo imaging probe that is able to demonstrate in vivo target engagement is critical and desirable. Brequinar is one of the most potent hDHODH inhibitors so far discovered. In this work, we use a copper-mediated radiofluorination (CMRF) strategy and compare the chemical design and radiosynthesis starting from either pinacole boronate p-nitrobenzyl ester (4) or tributylstannate (tin) p-nitrobenzyl ester (5), chosen for their suitability as a precursor to [18F]brequinar. We report here the design, synthesis, radiolabeling and characterization of [18F]brequinar, and a preliminary PET imaging study of DHODH in vivo. This study provides the strategies to create [18F]brequinar, the first hDHODH inhibitor PET radiotracer, which will facilitate its use as a tool (theranostics) for hDHODH drug development and for diagnosis and monitoring therapeutic efficacy in AML and cancers.
Collapse
Affiliation(s)
- Vinay Kumar Banka
- Department of Radiology, New York University School of Medicine, Center for Biomedical Imaging 660 First Ave., 4th Floor New York NY 10016 USA +1 (212)263 7541 +1 (212)263 6605
| | - Stefano Sainas
- Department of Drug Science and Technology (DSTF), University of Torino Turin Italy
| | - Elena Martino
- Department of Drug Science and Technology (DSTF), University of Torino Turin Italy
| | - Jiacheng Wang
- Department of Radiology, New York University School of Medicine, Center for Biomedical Imaging 660 First Ave., 4th Floor New York NY 10016 USA +1 (212)263 7541 +1 (212)263 6605
| | - Marco Lucio Lolli
- Department of Drug Science and Technology (DSTF), University of Torino Turin Italy
| | - Yu-Shin Ding
- Department of Radiology, New York University School of Medicine, Center for Biomedical Imaging 660 First Ave., 4th Floor New York NY 10016 USA +1 (212)263 7541 +1 (212)263 6605
- Department of Psychiatry, New York University School of Medicine New York NY USA
| |
Collapse
|
3
|
Gehlot P, Vyas VK. A Patent Review of Human Dihydroorotate Dehydrogenase (hDHODH) Inhibitors as Anticancer Agents and their Other Therapeutic Applications (1999-2022). Recent Pat Anticancer Drug Discov 2024; 19:280-297. [PMID: 37070439 DOI: 10.2174/1574892818666230417094939] [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: 11/15/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 04/19/2023]
Abstract
Highly proliferating cells, such as cancer cells, are in high demand of pyrimidine nucleotides for their proliferation, accomplished by de novo pyrimidine biosynthesis. The human dihydroorotate dehydrogenase (hDHODH) enzyme plays a vital role in the rate-limiting step of de novo pyrimidine biosynthesis. As a recognised therapeutic target, hDHODH plays a significant role in cancer and other illness. In the past two decades, small molecules as inhibitors hDHODH enzyme have drawn much attention as anticancer agents, and their role in rheumatoid arthritis (RA), and multiple sclerosis (MS). In this patent review, we have compiled patented hDHODH inhibitors published between 1999 and 2022 and discussed the development of hDHODH inhibitors as anticancer agents. Therapeutic potential of small molecules as hDHODH inhibitors for the treatment of various diseases, such as cancer, is very well recognised. Human DHODH inhibitors can rapidly cause intracellular uridine monophosphate (UMP) depletion to produce starvation of pyrimidine bases. Normal cells can better endure a brief period of starvation without the side effects of conventional cytotoxic medication and resume synthesis of nucleic acid and other cellular functions after inhibition of de novo pathway using an alternative salvage pathway. Highly proliferative cells such as cancer cells do not endure starvation because they are in high demand of nucleotides for cell differentiation, which is fulfilled by de novo pyrimidine biosynthesis. In addition, hDHODH inhibitors produce their desired activity at lower doses rather than a cytotoxic dose of other anticancer agents. Thus, inhibition of de novo pyrimidine biosynthesis will create new prospects for the development of novel targeted anticancer agents, which ongoing preclinical and clinical experiments define. Our work brings together a comprehensive patent review of the role of hDHODH in cancer, as well as various patents related to the hDHODH inhibitors and their anticancer and other therapeutic potential. This compiled work on patented DHODH inhibitors will guide researchers in pursuing the most promising drug discovery strategies against the hDHODH enzyme as anticancer agents.
Collapse
Affiliation(s)
- Pinky Gehlot
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujrat, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujrat, India
| |
Collapse
|
4
|
Vyas VK, Shukla T, Sharma M. Medicinal chemistry approaches for the discovery of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors as antimalarial agents. Future Med Chem 2023; 15:1295-1321. [PMID: 37551689 DOI: 10.4155/fmc-2023-0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Malaria is a severe human disease and a global health problem because of drug-resistant strains. Drugs reported to prevent the growth of Plasmodium parasites target various phases of the parasites' life cycle. Antimalarial drugs can inhibit key enzymes that are responsible for the cellular growth and development of parasites. Plasmodium falciparum dihydroorotate dehydrogenase is one such enzyme that is necessary for de novo pyrimidine biosynthesis. This review focuses on various medicinal chemistry approaches used for the discovery and identification of selective P. falciparum dihydroorotate dehydrogenase inhibitors as antimalarial agents. This comprehensive review discusses recent advances in the selective therapeutic activity of distinct chemical classes of compounds as P. falciparum dihydroorotate dehydrogenase inhibitors and antimalarial drugs.
Collapse
Affiliation(s)
- Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Tanvi Shukla
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| |
Collapse
|
5
|
Shibata T, Narita T, Suto Y, Yasmin H, Kabashima T. A Facile Fluorometric Assay of Orotate Phosphoribosyltransferase Activity Using a Selective Fluorogenic Reaction for Orotic Acid. SENSORS (BASEL, SWITZERLAND) 2023; 23:2507. [PMID: 36904710 PMCID: PMC10007123 DOI: 10.3390/s23052507] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 06/18/2023]
Abstract
Orotate phosphoribosyltransferase (OPRT) exists as a bifunctional enzyme, uridine 5'-monophosphate synthase, in mammalian cells and plays an important role in pyrimidine biosynthesis. Measuring OPRT activity has been considered important for understanding biological events and development of molecular-targeting drugs. In this study, we demonstrate a novel fluorescence method for measuring OPRT activity in living cells. The technique utilizes 4-trifluoromethylbenzamidoxime (4-TFMBAO) as a fluorogenic reagent, which produces selective fluorescence for orotic acid. To perform the OPRT reaction, orotic acid was added to HeLa cell lysate, and a portion of the enzyme reaction mixture was heated at 80 °C for 4 min in the presence of 4-TFMBAO under basic conditions. The resulting fluorescence was measured using a spectrofluorometer, which reflects the consumption of orotic acid by the OPRT. After optimization of the reaction conditions, the OPRT activity was successfully determined in 15 min of enzyme reaction time without further procedures such as purification of OPRT or deproteination for the analysis. The activity obtained was compatible with the value measured by the radiometric method with [3H]-5-FU as the substrate. The present method provides a reliable and facile measurement of OPRT activity and could be useful for a variety of research fields targeting pyrimidine metabolism.
Collapse
Affiliation(s)
- Takayuki Shibata
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi 371-8514, Japan
| | - Tomohiro Narita
- Department of Pharmaceutical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Yutaka Suto
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 37-1 Nakaoruimachi, Takasaki 370-0033, Japan
| | - Hasina Yasmin
- Department of Pharmacy, BRAC University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | - Tsutomu Kabashima
- Graduate School of Pharmaceutical Sciences, Nagasaki International University, 2825-7 Huis Ten Bosch, Sasebo 859-3298, Japan
| |
Collapse
|
6
|
Zhang X, Tan X, Wang P, Qin J. Application of Polypyrrole-Based Electrochemical Biosensor for the Early Diagnosis of Colorectal Cancer. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:674. [PMID: 36839042 PMCID: PMC9967576 DOI: 10.3390/nano13040674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
Although colorectal cancer (CRC) is easy to treat surgically and can be combined with postoperative chemotherapy, its five-year survival rate is still not optimistic. Therefore, developing sensitive, efficient, and compliant detection technology is essential to diagnose CRC at an early stage, providing more opportunities for effective treatment and intervention. Currently, the widely used clinical CRC detection methods include endoscopy, stool examination, imaging modalities, and tumor biomarker detection; among them, blood biomarkers, a noninvasive strategy for CRC screening, have shown significant potential for early diagnosis, prediction, prognosis, and staging of cancer. As shown by recent studies, electrochemical biosensors have attracted extensive attention for the detection of blood biomarkers because of their advantages of being cost-effective and having sound sensitivity, good versatility, high selectivity, and a fast response. Among these, nano-conductive polymer materials, especially the conductive polymer polypyrrole (PPy), have been broadly applied to improve sensing performance due to their excellent electrical properties and the flexibility of their surface properties, as well as their easy preparation and functionalization and good biocompatibility. This review mainly discusses the characteristics of PPy-based biosensors, their synthetic methods, and their application for the detection of CRC biomarkers. Finally, the opportunities and challenges related to the use of PPy-based sensors for diagnosing CRC are also discussed.
Collapse
|
7
|
Xu Z, Lu S, Liu X, Tang L, Liu Z, Cui J, Wang W, Lu W, Huang J. Drug repurposing of ilepcimide that ameliorates experimental autoimmune encephalomyelitis via restricting inflammatory response and oxidative stress. Toxicol Appl Pharmacol 2023; 458:116328. [PMID: 36455640 DOI: 10.1016/j.taap.2022.116328] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS) that remains incurable. Herein, we demonstrated that ilepcimide (Antiepilepsirine), an antiepileptic drug used for decades, protects mice from experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Our studies found that ilepcimide treatment effectively ameliorates demyelination, blood-brain barrier leakage and infiltration of CD4+ and CD8+ T cells in EAE mice. On the one hand, ilepcimide can inhibit dihydroorotate dehydrogenase (DHODH), an important therapeutic target for MS. Computer molecular docking, thermal shift and fluorescence quenching assay demonstrated the directly interaction between ilepcimide and DHODH. Accordingly, ilepcimide observably repressed T cell proliferation in mixed lymphocyte reaction (MLR) assay and concanavalin A (Con-A) model in a DHODH-dependent manner. On the other hand, ilepcimide exhibited neuroprotective effect possibly through activating NRF2 antioxidant pathway in mouse neural crest-derived Neuro2a cells. Collectively, our findings have revealed the therapeutic potential of ilepcimide in EAE mouse model via restricting inflammatory response and oxidative stress, offering a potential opportunity for repurposing existing drug ilepcimide for MS therapy.
Collapse
Affiliation(s)
- Zhaomin Xu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Sisi Lu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xi Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Lu Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zehui Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiayan Cui
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wanyan Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Jin Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
8
|
Miyake S, Masuda S. Inhibition of mitochondrial complex III or dihydroorotate dehydrogenase (DHODH) triggers formation of poly(A) + RNA foci adjacent to nuclear speckles following activation of ATM (ataxia telangiectasia mutated). RNA Biol 2022; 19:1244-1255. [PMID: 36412986 PMCID: PMC9683070 DOI: 10.1080/15476286.2022.2146919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Intracellular and intercellular signalling networks play an essential role in optimizing cellular homoeostasis and are thought to be partly reflected in nuclear mRNA dynamics. However, the regulation of nuclear mRNA dynamics by intracellular and intercellular signals remains largely unexplored, and research tools are lacking. Through an original screening based on the mRNA metabolic mechanism, we discovered that eight well-known inhibitors cause significant nuclear poly(A)+ RNA accumulation. Among these inhibitors, we discovered a new mRNA metabolic response in which the addition of antimycin A, an inhibitor of mitochondrial respiratory-chain complex III (complex III), resulted in a marked accumulation of poly(A)+ RNA near the nuclear speckles. Furthermore, dihydroorotate dehydrogenase (DHODH) inhibitors, a rate-limiting enzyme in the intracellular de novo pyrimidine synthesis reaction that specifically exchanges electrons with complex III, also caused a remarkable accumulation of nuclear poly(A)+ RNA adjacent to the nuclear speckles, which was abolished by extracellular uridine supply, indicating that the depletion of intracellular pyrimidine affects poly(A)+ RNA metabolism. Further analysis revealed that ataxia telangiectasia mutated (ATM), a serine and threonine kinase and a master regulator of DNA double-strand break (DSB) and nucleolar stress, is required for this poly(A)+ RNA nuclear accumulation phenomenon. This study reports new insights into novel aspects of nuclear poly(A)+ RNA metabolism, especially the relationship between mitochondrial respiratory-chain functions, pyrimidine metabolism, and nuclear RNA metabolism.
Collapse
Affiliation(s)
- Shuntaro Miyake
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Seiji Masuda
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto, Japan,Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara, Japan,Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan,Antiaging Center, Kindai University, Higashiosaka, Japan,CONTACT Seiji Masuda Department of Food Science and Nutrition, Faculty of Agriculture, Kindai University, Nara631-8505, Japan
| |
Collapse
|
9
|
Zhou Y, Tao L, Zhou X, Zuo Z, Gong J, Liu X, Zhou Y, Liu C, Sang N, Liu H, Zou J, Gou K, Yang X, Zhao Y. DHODH and cancer: promising prospects to be explored. Cancer Metab 2021; 9:22. [PMID: 33971967 PMCID: PMC8107416 DOI: 10.1186/s40170-021-00250-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/10/2021] [Indexed: 02/08/2023] Open
Abstract
Human dihydroorotate dehydrogenase (DHODH) is a flavin-dependent mitochondrial enzyme catalyzing the fourth step in the de novo pyrimidine synthesis pathway. It is originally a target for the treatment of the non-neoplastic diseases involving in rheumatoid arthritis and multiple sclerosis, and is re-emerging as a validated therapeutic target for cancer therapy. In this review, we mainly unravel the biological function of DHODH in tumor progression, including its crucial role in de novo pyrimidine synthesis and mitochondrial respiratory chain in cancer cells. Moreover, various DHODH inhibitors developing in the past decades are also been displayed, and the specific mechanism between DHODH and its additional effects are illustrated. Collectively, we detailly discuss the association between DHODH and tumors in recent years here, and believe it will provide significant evidences and potential strategies for utilizing DHODH as a potential target in preclinical and clinical cancer therapies.
Collapse
Affiliation(s)
- Yue Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lei Tao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xia Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zeping Zuo
- The Laboratory of Anesthesiology and Critical Care Medicine, Translational Neuroscience Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jin Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaocong Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yang Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Chunqi Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Sang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Huan Liu
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jiao Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kun Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaowei Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yinglan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Qiu X, Jiang S, Xiao Y, He Y, Ren T, Jiang L, Liu R, Chen Q. SOX2-dependent expression of dihydroorotate dehydrogenase regulates oral squamous cell carcinoma cell proliferation. Int J Oral Sci 2021; 13:3. [PMID: 33510132 PMCID: PMC7844284 DOI: 10.1038/s41368-020-00109-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 02/08/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) become a heavy burden of public health, with approximately 300 000 newly diagnosed cases and 145 000 deaths worldwide per year. Nucleotide metabolism fuel DNA replication and RNA synthesis, which is indispensable for cell proliferation. But how tumor cells orchestrate nucleotide metabolic enzymes to support their rapid growth is largely unknown. Here we show that expression of pyrimidine metabolic enzyme dihydroorotate dehydrogenase (DHODH) is upregulated in OSCC tissues, compared to non-cancerous adjacent tissues. Enhanced expression of DHODH is correlated with a shortened patient survival time. Inhibition of DHODH by either shRNA or selective inhibitors impairs proliferation of OSCC cells and growth of tumor xenograft. Further, loss of functional DHODH imped de novo pyrimidine synthesis, and disrupt mitochondrial respiration probably through destabilizing the MICOS complex. Mechanistic study shows that transcriptional factor SOX2 plays an important role in the upregulation of DHODH in OSCC. Our findings add to the knowledge of how cancer cells co-opt nucleotide metabolism to support their rapid growth, and thereby highlight DHODH as a potential prognostic and therapeutic target for OSCC treatment.
Collapse
Affiliation(s)
- Xuemei Qiu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sheng Jiang
- Ministry of science and technology, The Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu, China
| | - Yanxuan Xiao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yumin He
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tao Ren
- Oncology Department, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Lu Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Rui Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Qian Y, Liang X, Kong P, Cheng Y, Cui H, Yan T, Wang J, Zhang L, Liu Y, Guo S, Cheng X, Cui Y. Elevated DHODH expression promotes cell proliferation via stabilizing β-catenin in esophageal squamous cell carcinoma. Cell Death Dis 2020; 11:862. [PMID: 33060568 PMCID: PMC7566478 DOI: 10.1038/s41419-020-03044-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 12/24/2022]
Abstract
As a key enzyme in de novo pyrimidine biosynthesis, the expression level of dihydroorotate dehydrogenase (DHODH) has been reported to be elevated in various types of malignant tumors and its tumor-promoting effect was considered to relate to its pyrimidine synthesis function. Here, we revealed one intriguing potential mechanism that DHODH modulated β-catenin signaling in esophageal squamous cell carcinoma (ESCC). We demonstrated that DHODH directly bound to the NH2 terminal of β-catenin, thereby, interrupting the interaction of GSK3β with β-catenin and leading to the abrogation of β-catenin degradation and accumulation of β-catenin in the nucleus, which in turn, resulted in the activation of β-catenin downstream genes, including CCND1, E2F3, Nanog, and OCT4. We further demonstrated that the regulation of β-catenin by DHODH was independent of DHODH catalyzing activity. Univariate and multivariate analyses suggested that DHODH expression might be an independent prognostic factor for ESCC patients. Collectively, our study highlights the pivotal role of DHODH mediated β-catenin signaling and indicates that DHODH may act as a multi-functional switcher from catalyzing pyrimidine metabolism to regulating tumor-related signaling pathways in ESCC.
Collapse
Affiliation(s)
- Yu Qian
- Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, 518035, Shenzhen, People's Republic of China.,Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, 030001, Taiyuan, Shanxi, People's Republic of China
| | - Xiao Liang
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, 030001, Taiyuan, Shanxi, People's Republic of China
| | - Pengzhou Kong
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, 030001, Taiyuan, Shanxi, People's Republic of China
| | - Yikun Cheng
- Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, 518035, Shenzhen, People's Republic of China
| | - Heyang Cui
- Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, 518035, Shenzhen, People's Republic of China.,Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, 030001, Taiyuan, Shanxi, People's Republic of China
| | - Ting Yan
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, 030001, Taiyuan, Shanxi, People's Republic of China
| | - Jinghao Wang
- Department of Obstetrics & Gynecology, the Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ling Zhang
- Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, 518035, Shenzhen, People's Republic of China.,Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, 030001, Taiyuan, Shanxi, People's Republic of China
| | - Yiqian Liu
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, 030001, Taiyuan, Shanxi, People's Republic of China
| | - Shiping Guo
- Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Xiaolong Cheng
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, 030001, Taiyuan, Shanxi, People's Republic of China.
| | - Yongping Cui
- Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, 518035, Shenzhen, People's Republic of China.
| |
Collapse
|
12
|
Liu Z, Hu Q, Wang W, Lu S, Wu D, Ze S, He J, Huang Y, Chen W, Xu Y, Lu W, Huang J. Natural product piperine alleviates experimental allergic encephalomyelitis in mice by targeting dihydroorotate dehydrogenase. Biochem Pharmacol 2020; 177:114000. [PMID: 32353424 DOI: 10.1016/j.bcp.2020.114000] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/24/2020] [Indexed: 01/06/2023]
Abstract
Multiple sclerosis (MS) is the most popular chronic and debilitating inflammatory disease of the central nervous system (CNS) that remains incurable. Dihydroorotate dehydrogenase (DHODH) is critical to the activity of T lymphocytes and represents a potential therapeutic target for MS. Here we identify piperine, a bioactive constituent of black pepper, as a potent inhibitor of DHODH with an IC50 value of 0.88 μM. Isothermal titration calorimetry and thermofluor assay demonstrate the directly interaction between piperine and DHODH. The co-complex crystal structure of DHODH and piperine at 1.98 Å resolution further reveal that Tyr356 residue of DHODH is crucial for piperine binding. Importantly, we show that piperine can inhibit T cell overactivation in a DHODH-dependent manner in concanavalin A-triggered T-cell assay and mixed lymphocyte reaction assay. Finally, piperine exhibits strong preventive and therapeutic effect in the MOG-induced experimental allergic encephalomyelitis (EAE), a useful model for studying potential treatments for MS, by restricting inflammatory cells infiltration into the CNS and preventing myelin destruction and blood-brain barrier (BBB) disruption. Taken together, these findings highlight DHODH as a therapeutic target for autoimmune disease of the nervous system, and demonstrate a novel role for piperine in the treatment of MS.
Collapse
Affiliation(s)
- Zehui Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Qian Hu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wanyan Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Sisi Lu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Dang Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shuyin Ze
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiacheng He
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ying Huang
- Guangdong Institute for Drug Control, Guangdong, China
| | - Wuyan Chen
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Yechun Xu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
13
|
Boukalova S, Hubackova S, Milosevic M, Ezrova Z, Neuzil J, Rohlena J. Dihydroorotate dehydrogenase in oxidative phosphorylation and cancer. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165759. [PMID: 32151633 DOI: 10.1016/j.bbadis.2020.165759] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
Dihydroorotate dehydrogenase (DHODH) is an enzyme of the de novo pyrimidine synthesis pathway that provides nucleotides for RNA/DNA synthesis essential for proliferation. In mammalian cells, DHODH is localized in mitochondria, linked to the respiratory chain via the coenzyme Q pool. Here we discuss the role of DHODH in the oxidative phosphorylation system and in the initiation and progression of cancer. We summarize recent findings on DHODH biology, the progress made in the development of new, specific inhibitors of DHODH intended for cancer therapy, and the mechanistic insights into the consequences of DHODH inhibition.
Collapse
Affiliation(s)
- Stepana Boukalova
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 252 50 Vestec, Prague-West, Czech Republic
| | - Sona Hubackova
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 252 50 Vestec, Prague-West, Czech Republic
| | - Mirko Milosevic
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 252 50 Vestec, Prague-West, Czech Republic
| | - Zuzana Ezrova
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 252 50 Vestec, Prague-West, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 252 50 Vestec, Prague-West, Czech Republic; School of Medical Science, Griffith University, Southport, 4222, Qld, Australia
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 252 50 Vestec, Prague-West, Czech Republic.
| |
Collapse
|
14
|
Hubackova S, Davidova E, Boukalova S, Kovarova J, Bajzikova M, Coelho A, Terp MG, Ditzel HJ, Rohlena J, Neuzil J. Replication and ribosomal stress induced by targeting pyrimidine synthesis and cellular checkpoints suppress p53-deficient tumors. Cell Death Dis 2020; 11:110. [PMID: 32034120 PMCID: PMC7007433 DOI: 10.1038/s41419-020-2224-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/22/2022]
Abstract
p53-mutated tumors often exhibit increased resistance to standard chemotherapy and enhanced metastatic potential. Here we demonstrate that inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme of the de novo pyrimidine synthesis pathway, effectively decreases proliferation of cancer cells via induction of replication and ribosomal stress in a p53- and checkpoint kinase 1 (Chk1)-dependent manner. Mechanistically, a block in replication and ribosomal biogenesis result in p53 activation paralleled by accumulation of replication forks that activate the ataxia telangiectasia and Rad3-related kinase/Chk1 pathway, both of which lead to cell cycle arrest. Since in the absence of functional p53 the cell cycle arrest fully depends on Chk1, combined DHODH/Chk1 inhibition in p53-dysfunctional cancer cells induces aberrant cell cycle re-entry and erroneous mitosis, resulting in massive cell death. Combined DHODH/Chk1 inhibition effectively suppresses p53-mutated tumors and their metastasis, and therefore presents a promising therapeutic strategy for p53-mutated cancers.
Collapse
Affiliation(s)
- Sona Hubackova
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, 252 50, Czech Republic.
| | - Eliska Davidova
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, 252 50, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Stepana Boukalova
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, 252 50, Czech Republic
| | - Jaromira Kovarova
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, 252 50, Czech Republic
| | - Martina Bajzikova
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, 252 50, Czech Republic
| | - Ana Coelho
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, 252 50, Czech Republic
| | - Mikkel G Terp
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Department of Oncology, Odense University Hospital, 5000, Odense, Denmark
| | - Jakub Rohlena
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, 252 50, Czech Republic
| | - Jiri Neuzil
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, 252 50, Czech Republic. .,School of Medical Science, Griffith University, Southport, QLD, 4222, Australia.
| |
Collapse
|
15
|
HIF1α inhibition facilitates Leflunomide-AHR-CRP signaling to attenuate bone erosion in CRP-aberrant rheumatoid arthritis. Nat Commun 2019; 10:4579. [PMID: 31594926 PMCID: PMC6783548 DOI: 10.1038/s41467-019-12163-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 08/16/2019] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disorder characterized by progressive bone erosion. Leflunomide is originally developed to suppress inflammation via its metabolite A77 1726 to attenuate bone erosion. However, distinctive responsiveness to Leflunomide is observed among RA individuals. Here we show that Leflunomide exerts immunosuppression but limited efficacy in RA individuals distinguished by higher serum C-reactive protein (CRPHigher, CRPH), whereas the others with satisfactory responsiveness to Leflunomide show lower CRP (CRPLower, CRPL). CRP inhibition decreases bone erosion in arthritic rats. Besides the immunomodulation via A77 1726, Leflunomide itself induces AHR-ARNT interaction to inhibit hepatic CRP production and attenuate bone erosion in CRPL arthritic rats. Nevertheless, high CRP in CRPH rats upregulates HIF1α, which competes with AHR for ARNT association and interferes Leflunomide-AHR-CRP signaling. Hepatocyte-specific HIF1α deletion or a HIF1α inhibitor Acriflavine re-activates Leflunomide-AHR-CRP signaling to inhibit bone erosion. This study presents a precision medicine-based therapeutic strategy for RA. Leflunomide is used for the treatment of rheumatoid arthritis. Here, the authors show that effectiveness is limited in patients with higher levels of serum c-reactive protein (CRP). Using animal models, they show that higher CRP induces HIF1a expression, which in turn interferes with Leflunomide signalling, and that effectiveness of the drug is restored when HIF1a is pharmacologically inhibited.
Collapse
|
16
|
Bajzikova M, Kovarova J, Coelho AR, Boukalova S, Oh S, Rohlenova K, Svec D, Hubackova S, Endaya B, Judasova K, Bezawork-Geleta A, Kluckova K, Chatre L, Zobalova R, Novakova A, Vanova K, Ezrova Z, Maghzal GJ, Magalhaes Novais S, Olsinova M, Krobova L, An YJ, Davidova E, Nahacka Z, Sobol M, Cunha-Oliveira T, Sandoval-Acuña C, Strnad H, Zhang T, Huynh T, Serafim TL, Hozak P, Sardao VA, Koopman WJH, Ricchetti M, Oliveira PJ, Kolar F, Kubista M, Truksa J, Dvorakova-Hortova K, Pacak K, Gurlich R, Stocker R, Zhou Y, Berridge MV, Park S, Dong L, Rohlena J, Neuzil J. Reactivation of Dihydroorotate Dehydrogenase-Driven Pyrimidine Biosynthesis Restores Tumor Growth of Respiration-Deficient Cancer Cells. Cell Metab 2019; 29:399-416.e10. [PMID: 30449682 PMCID: PMC7484595 DOI: 10.1016/j.cmet.2018.10.014] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 08/04/2018] [Accepted: 10/24/2018] [Indexed: 12/29/2022]
Abstract
Cancer cells without mitochondrial DNA (mtDNA) do not form tumors unless they reconstitute oxidative phosphorylation (OXPHOS) by mitochondria acquired from host stroma. To understand why functional respiration is crucial for tumorigenesis, we used time-resolved analysis of tumor formation by mtDNA-depleted cells and genetic manipulations of OXPHOS. We show that pyrimidine biosynthesis dependent on respiration-linked dihydroorotate dehydrogenase (DHODH) is required to overcome cell-cycle arrest, while mitochondrial ATP generation is dispensable for tumorigenesis. Latent DHODH in mtDNA-deficient cells is fully activated with restoration of complex III/IV activity and coenzyme Q redox-cycling after mitochondrial transfer, or by introduction of an alternative oxidase. Further, deletion of DHODH interferes with tumor formation in cells with fully functional OXPHOS, while disruption of mitochondrial ATP synthase has little effect. Our results show that DHODH-driven pyrimidine biosynthesis is an essential pathway linking respiration to tumorigenesis, pointing to inhibitors of DHODH as potential anti-cancer agents.
Collapse
Affiliation(s)
- Martina Bajzikova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic; Faculty of Science, Charles University, 128 44 Prague, Czech Republic
| | - Jaromira Kovarova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic.
| | - Ana R Coelho
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Stepana Boukalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Sehyun Oh
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul 08826, Korea
| | - Katerina Rohlenova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - David Svec
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Sona Hubackova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Berwini Endaya
- School of Medical Science, Griffith University, Southport, QLD 4222, Australia
| | - Kristyna Judasova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | | | - Katarina Kluckova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Laurent Chatre
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015 Paris, France; CNRS UMR 3738, Team Stability of Nuclear and Mitochondrial DNA, 75015 Paris, France
| | - Renata Zobalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Anna Novakova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Katerina Vanova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Zuzana Ezrova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic; Faculty of Science, Charles University, 128 44 Prague, Czech Republic
| | - Ghassan J Maghzal
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, UNSW Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Silvia Magalhaes Novais
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic; Faculty of Science, Charles University, 128 44 Prague, Czech Republic
| | - Marie Olsinova
- Faculty of Science, Charles University, 128 44 Prague, Czech Republic
| | - Linda Krobova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Yong Jin An
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul 08826, Korea
| | - Eliska Davidova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic; Faculty of Science, Charles University, 128 44 Prague, Czech Republic
| | - Zuzana Nahacka
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Margarita Sobol
- Institute of Molecular Genetics, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Teresa Cunha-Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Cristian Sandoval-Acuña
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Hynek Strnad
- Institute of Molecular Genetics, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Tongchuan Zhang
- Institute for Glycomics, Griffith University, Southport, 4222 QLD, Australia
| | - Thanh Huynh
- Eunice Kennedy Shriver Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Teresa L Serafim
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Pavel Hozak
- Institute of Molecular Genetics, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Vilma A Sardao
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Werner J H Koopman
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 Nijmegen, the Netherlands
| | - Miria Ricchetti
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015 Paris, France; CNRS UMR 3738, Team Stability of Nuclear and Mitochondrial DNA, 75015 Paris, France
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Frantisek Kolar
- Institute of Physiology, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Mikael Kubista
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Jaroslav Truksa
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic
| | - Katerina Dvorakova-Hortova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic; Faculty of Science, Charles University, 128 44 Prague, Czech Republic
| | - Karel Pacak
- Eunice Kennedy Shriver Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Robert Gurlich
- Third Faculty Hospital, Charles University, Prague, Czech Republic
| | - Roland Stocker
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, UNSW Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yaoqi Zhou
- Institute for Glycomics, Griffith University, Southport, 4222 QLD, Australia
| | | | - Sunghyouk Park
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul 08826, Korea.
| | - Lanfeng Dong
- School of Medical Science, Griffith University, Southport, QLD 4222, Australia.
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic.
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Vestec, Prague-West, Czech Republic; School of Medical Science, Griffith University, Southport, QLD 4222, Australia.
| |
Collapse
|
17
|
Identification of New Activators of Mitochondrial Fusion Reveals a Link between Mitochondrial Morphology and Pyrimidine Metabolism. Cell Chem Biol 2017; 25:268-278.e4. [PMID: 29290623 DOI: 10.1016/j.chembiol.2017.12.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/12/2017] [Accepted: 11/30/2017] [Indexed: 01/26/2023]
Abstract
Mitochondria are dynamic organelles that produce most of the cellular ATP, and are involved in many other cellular functions such as Ca2+ signaling, differentiation, apoptosis, cell cycle, and cell growth. One key process of mitochondrial dynamics is mitochondrial fusion, which is catalyzed by mitofusins (MFN1 and MFN2) and OPA1. The outer mitochondrial membrane protein MFN2 plays a relevant role in the maintenance of mitochondrial metabolism, insulin signaling, and mutations that cause neurodegenerative disorders. Therefore, modulation of proteins involved in mitochondrial dynamics has emerged as a potential pharmacological strategy. Here, we report the identification of small molecules by high-throughput screen that promote mitochondrial elongation in an MFN1/MFN2-dependent manner. Detailed analysis of their mode of action reveals a previously unknown connection between pyrimidine metabolism and mitochondrial dynamics. Our data indicate a link between pyrimidine biosynthesis and mitochondrial dynamics, which maintains cell survival under stress conditions characterized by loss of pyrimidine synthesis.
Collapse
|
18
|
Madak JT, Cuthbertson CR, Chen W, Showalter HD, Neamati N. Design, Synthesis, and Characterization of Brequinar Conjugates as Probes to Study DHODH Inhibition. Chemistry 2017; 23:13875-13878. [DOI: 10.1002/chem.201702999] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Joseph T. Madak
- Department of Medicinal Chemistry, College of Pharmacy, and Translational Oncology Program; University of Michigan, North Campus Research Complex; 1600 Huron Parkway Ann Arbor MI 48109 USA
| | - Christine R. Cuthbertson
- Department of Medicinal Chemistry, College of Pharmacy, and Translational Oncology Program; University of Michigan, North Campus Research Complex; 1600 Huron Parkway Ann Arbor MI 48109 USA
| | - Wenmin Chen
- Department of Medicinal Chemistry, College of Pharmacy, and Translational Oncology Program; University of Michigan, North Campus Research Complex; 1600 Huron Parkway Ann Arbor MI 48109 USA
| | - Hollis D. Showalter
- Department of Medicinal Chemistry, College of Pharmacy; University of Michigan; 428 Church St. Ann Arbor MI 48109 USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, and Translational Oncology Program; University of Michigan, North Campus Research Complex; 1600 Huron Parkway Ann Arbor MI 48109 USA
| |
Collapse
|
19
|
Liu B, Fu XQ, Li T, Su T, Guo H, Zhu PL, Tse AKW, Liu SM, Yu ZL. Computational and experimental prediction of molecules involved in the anti-melanoma action of berberine. JOURNAL OF ETHNOPHARMACOLOGY 2017; 208:225-235. [PMID: 28729227 DOI: 10.1016/j.jep.2017.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 05/07/2017] [Accepted: 07/15/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGIC RELEVANCE Berberine (BBR) is a naturally occurring alkaloid compound that can be found in Chinese medicinal herbs such as Rhizoma Coptidis and Phellodendri Cortex. These BBR containing herbs are commonly used by Chinese medicine doctors to treat cancers including melanoma. In this study, we explored proteins potentially involved in the anti-melanoma effects of BBR using computational and experimental approaches. MATERIALS AND METHODS Target proteins of BBR were predicted using the reverse pharmacophore screening, molecular docking and molecular dynamics. Anti-melanoma activities of BBR in melanoma cells were examined by MTT and EdU proliferation assays. Effects of BBR on activities of target proteins in melanoma cells were examined by Western blotting or fluorescence assay. RESULTS Ten proteins implicated in cancer and with high fit-score in the reverse pharmacophore screening were selected as potential targets of BBR. Molecular docking and molecular dynamics revealed that BBR could stably bind to four of the ten proteins, namely 3-phosphoinositide-dependent protein kinase 1 (PDK1), glucocorticoid receptor (GR), p38 mitogen-activated protein kinase (p38) and dihydroorotate dehydrogenase (DHODH). Cellular experiments showed that BBR inhibited cell proliferation, increased the phosphorylation of GR and p38, and inhibited the activity of DHODH in A375 human melanoma cells. CONCLUSIONS These findings suggest that p38, GR and DHODH are potentially involved in the anti-melanoma action of BBR. This study provided a chemical and pharmacological justification for the clinical use of BBR-containing herbs in melanoma treatment.
Collapse
Affiliation(s)
- Bin Liu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China; Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiu-Qiong Fu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Ting Li
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Tao Su
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Hui Guo
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Pei-Li Zhu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Anfernee Kai-Wing Tse
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Shi-Ming Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Zhi-Ling Yu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China; HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.
| |
Collapse
|
20
|
Reis RAG, Calil FA, Feliciano PR, Pinheiro MP, Nonato MC. The dihydroorotate dehydrogenases: Past and present. Arch Biochem Biophys 2017; 632:175-191. [PMID: 28666740 DOI: 10.1016/j.abb.2017.06.019] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 01/24/2023]
Abstract
The flavoenzyme dihydroorotate dehydrogenase catalyzes the stereoselective oxidation of (S)-dihydroorotate to orotate in the fourth of the six conserved enzymatic reactions involved in the de novo pyrimidine biosynthetic pathway. Inhibition of pyrimidine metabolism by selectively targeting DHODHs has been exploited in the development of new therapies against cancer, immunological disorders, bacterial and viral infections, and parasitic diseases. Through a chronological narrative, this review summarizes the efforts of the scientific community to achieve our current understanding of structural and biochemical properties of DHODHs. It also attempts to describe the latest advances in medicinal chemistry for therapeutic development based on the selective inhibition of DHODH, including an overview of the experimental techniques used for ligand screening during the process of drug discovery.
Collapse
Affiliation(s)
- Renata A G Reis
- Department of Chemistry, Georgia State University, Atlanta, GA 30302, United States
| | - Felipe Antunes Calil
- Laboratório de Cristalografia de Proteínas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, 14040-903, Brazil
| | - Patricia Rosa Feliciano
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Matheus Pinto Pinheiro
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil
| | - M Cristina Nonato
- Laboratório de Cristalografia de Proteínas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, 14040-903, Brazil.
| |
Collapse
|