1
|
Lin F, Li J, Zhou L, Yi R, Chen Y, He S. Targeting vulnerability in tumor therapy: Dihydroorotate dehydrogenase. Life Sci 2025; 371:123612. [PMID: 40187643 DOI: 10.1016/j.lfs.2025.123612] [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: 01/16/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Dihydroorotate dehydrogenase (DHODH) is a key enzyme in the de novo pyrimidine biosynthetic pathway and a recognized therapeutic target in various diseases. In oncology research, DHODH has gained increasing importance and become a hot target for various tumor therapy studies. This review highlights three key points: (1) DHODH enables its diverse biological functions through its unique structural features and dominates the regulation of tumor metabolism and cell fate; (2) DHODH activates oncogenic signals, drives metastatic adaptation, and remodels drug resistance networks in tumors, making it a metabolic-signaling dual hub; and (3) DHODH inhibitors have shown significant efficacy in preclinical models of various tumors but face multiple challenges in clinical trials, including drug-related limitations and external constraints. Given these challenges, future research should explore DHODH inhibitors as a foundation for overcoming technological and translational barriers while establishing a systematic framework for the clinical application of DHODH-targeted tumor therapies.
Collapse
Affiliation(s)
- Fu Lin
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Jiaxin Li
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Lei Zhou
- Laboratory of Pathogen Biology and Immunology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Rigui Yi
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Yingge Chen
- School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Shuai He
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China.
| |
Collapse
|
2
|
Chen C, Wang L, Cui XF, Shang XY, Bai SH, Li L, Wang N, Han ZG. SCARA5 deficiency inhibits ferroptosis via regulating iron homeostasis and results in sorafenib resistance in hepatocellular carcinoma. Cell Signal 2025; 129:111656. [PMID: 39954713 DOI: 10.1016/j.cellsig.2025.111656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
SCARA5 (Scavenger Receptor Class A Member 5), a member of scavenger receptor class A, is a type II transmembrane protein. Previous studies, including our own, have suggested that SCARA5 acts as a tumor suppressor in various cancers. Additionally, SCARA5 has been identified as a ferritin receptor that facilitates iron delivery independent of transferrin. However, it remains unclear whether ferroptosis is involved in the tumor-suppressive function of SCARA5 in hepatocellular carcinoma (HCC). In this study, we found that SCARA5-deficient cells, including mouse embryonic fibroblasts (MEFs) and HCC cells, exhibited reduced sensitivity to ferroptosis induced by erastin and RSL3. We measured the cell viability, cellular reactive oxygen species (ROS), lipid ROS, malondialdehyde (MDA) and ferrous iron concentration to assess the role of SCARA5 in ferroptosis. Mechanistically, we confirmed that SCARA5 might enhance the intracellular availability of bioactive ferrous iron by promoting autophagic degradation of the major iron storage protein ferritin. Furthermore, we found that SCARA5 deficiency contributed to the resistance of HCC cells to sorafenib, a therapeutic agent for HCC, possibly by inhibiting ferroptosis. Collectively, our study revealed the role of SCARA5 in regulating ferroptosis, providing a profound understanding of sorafenib resistance in HCC systemic therapy.
Collapse
Affiliation(s)
- Cong Chen
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lan Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiao-Fang Cui
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xu-Yang Shang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shi-Hao Bai
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lin Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Na Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ze-Guang Han
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
3
|
Meijer T, Ter Braak B, Loonstra-Wolters L, Kunnen SJ, Islam B, Suciu I, Gardner I, Hatley O, Currie R, Hardy B, Leist M, van de Water B, Jennings P, Wilmes A. Transcriptomic changes and mitochondrial toxicity in response to acute and repeat dose treatment with brequinar in human liver and kidney in vitro models. Toxicol In Vitro 2025; 104:106010. [PMID: 39900124 DOI: 10.1016/j.tiv.2025.106010] [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: 11/04/2024] [Revised: 12/19/2024] [Accepted: 01/17/2025] [Indexed: 02/05/2025]
Abstract
The potent dihydroorotate dehydrogenase (DHODH) inhibitor brequinar has been investigated as an anticancer, immunosuppressive, and antiviral pharmaceutical agent. However, its toxicity is still poorly understood. We investigated the cellular responses of primary human hepatocytes (PHH) and telomerase-immortalised human renal proximal tubular epithelial cells (RPTEC/TERT1) after a single 24-h exposure up to 100 μM brequinar. Additionally, RPTEC/TERT1 cells underwent repeated daily exposure for five consecutive days at 0.3, 3, and 20 μM. Transcriptomic analysis revealed that PHH were less sensitive to brequinar treatment than RPTEC/TERT1 cells. Upregulation of various phase I and II drug-metabolising enzymes, particularly Cytochrome P450 (CYP) 1 A and 3 A enzymes, in PHH suggests potential detoxification. Furthermore, brequinar exposure led to a significant upregulation of several stress response pathways in PHH and RPTEC/TERT1 cells, including the unfolded protein response, Nrf2, p53, and inflammatory responses. RPTEC/TERT1 cells exhibited greater sensitivity to brequinar at 0.3 μM with repeated exposure compared to a single exposure. Furthermore, brequinar could impair the mitochondrial respiration of RPTEC/TERT1 cells after 24 h. This study provides new insights into the differential responses of PHH and RPTEC/TERT1 cells in response to brequinar exposure and highlights the biological relevance of implementing repeated dosing regimens in in vitro studies.
Collapse
Affiliation(s)
- Tamara Meijer
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands.
| | - Bas Ter Braak
- Cell Systems and Drug Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands; Toxys B.V., Leiden Bioscience Park, 2342 DH Oegstgeest, the Netherlands
| | - Liesanne Loonstra-Wolters
- Cell Systems and Drug Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands; Toxys B.V., Leiden Bioscience Park, 2342 DH Oegstgeest, the Netherlands
| | - Steven J Kunnen
- Cell Systems and Drug Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| | - Barira Islam
- Certara - Simcyp Division, Sheffield, United Kingdom.
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78464 Konstanz, Germany
| | - Iain Gardner
- Certara - Simcyp Division, Sheffield, United Kingdom
| | - Oliver Hatley
- Certara - Simcyp Division, Sheffield, United Kingdom
| | - Richard Currie
- Syngenta Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, UK
| | - Barry Hardy
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland.
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78464 Konstanz, Germany
| | - Bob van de Water
- Cell Systems and Drug Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| | - Paul Jennings
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands.
| | - Anja Wilmes
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Pham BQ, Yi SA, Ordureau A, An H. mTORC1 regulates the pyrimidine salvage pathway by controlling UCK2 turnover via the CTLH-WDR26 E3 ligase. Cell Rep 2025; 44:115179. [PMID: 39808525 PMCID: PMC11840829 DOI: 10.1016/j.celrep.2024.115179] [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: 08/29/2024] [Revised: 11/20/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
One critical aspect of cell proliferation is increased nucleotide synthesis, including pyrimidines. Pyrimidines are synthesized through de novo and salvage pathways. Prior studies established that the mammalian target of rapamycin complex 1 (mTORC1) promotes pyrimidine synthesis by activating the de novo pathway for cell proliferation. However, the involvement of mTORC1 in regulating the salvage pathway remains unclear. Here, we report that mTORC1 controls the half-life of uridine cytidine kinase 2 (UCK2), the rate-limiting enzyme in the salvage pathway. Specifically, UCK2 is degraded via the CTLH-WDR26 E3 complex during mTORC1 inhibition, which is prevented when mTORC1 is active. We also find that UCK1, an isoform of UCK2, affects the turnover of UCK2 by influencing its cellular localization. Importantly, altered UCK2 levels through the mTORC1-CTLH E3 pathway affect pyrimidine salvage and the efficacy of pyrimidine analog prodrugs. Therefore, mTORC1-CTLH E3-mediated degradation of UCK2 adds another layer of complexity to mTORC1's role in regulating pyrimidine metabolism.
Collapse
Affiliation(s)
- Brittany Q Pham
- Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sang Ah Yi
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heeseon An
- Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA; Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Tri-Institutional PhD Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
5
|
Maharati A, Rajabloo Y, Moghbeli M. Molecular mechanisms of mTOR-mediated cisplatin response in tumor cells. Heliyon 2025; 11:e41483. [PMID: 39834411 PMCID: PMC11743095 DOI: 10.1016/j.heliyon.2024.e41483] [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] [Received: 10/28/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025] Open
Abstract
Cisplatin (CDDP) is one of the main chemotherapeutic drugs that is widely used in many cancers. However, CDDP resistance is a frequent therapeutic challenge that reduces prognosis in cancer patients. Since, CDDP has noticeable side effects in normal tissues and organs, it is necessary to assess the molecular mechanisms associated with CDDP resistance to improve the therapeutic methods in cancer patients. Drug efflux, detoxifying systems, DNA repair mechanisms, and drug-induced apoptosis are involved in multidrug resistance in CDDP-resistant tumor cells. Mammalian target of rapamycin (mTOR), as a serine/threonine kinase has a pivotal role in various cellular mechanisms such as autophagy, metabolism, drug efflux, and cell proliferation. Although, mTOR is mainly activated by PI3K/AKT pathway, it can also be regulated by many other signaling pathways. PI3K/Akt/mTOR axis functions as a key modulator of drug resistance and unfavorable prognosis in different cancers. Regarding, the pivotal role of mTOR in CDDP response, in the present review we discussed the molecular mechanisms that regulate mTOR mediated CDDP response in tumor cells.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yasamin Rajabloo
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Meng J, Zhang L, He Z, Hu M, Liu J, Bao W, Tian Q, Feng H, Liu H. Development of a machine learning-based target-specific scoring function for structure-based binding affinity prediction for human dihydroorotate dehydrogenase inhibitors. J Comput Chem 2025; 46:e27510. [PMID: 39325045 DOI: 10.1002/jcc.27510] [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: 07/08/2024] [Revised: 08/21/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
Human dihydroorotate dehydrogenase (hDHODH) is a flavin mononucleotide-dependent enzyme that can limit de novo pyrimidine synthesis, making it a therapeutic target for diseases such as autoimmune disorders and cancer. In this study, using the docking structures of complexes generated by AutoDock Vina, we integrate interaction features and ligand features, and employ support vector regression to develop a target-specific scoring function for hDHODH (TSSF-hDHODH). The Pearson correlation coefficient values of TSSF-hDHODH in the cross-validation and external validation are 0.86 and 0.74, respectively, both of which are far superior to those of classic scoring function AutoDock Vina and random forest (RF) based generic scoring function RF-Score. TSSF-hDHODH is further used for the virtual screening of potential inhibitors in the FDA-Approved & Pharmacopeia Drug Library. In conjunction with the results from molecular dynamics simulations, crizotinib is identified as a candidate for subsequent structural optimization. This study can be useful for the discovery of hDHODH inhibitors and the development of scoring functions for additional targets.
Collapse
Affiliation(s)
- Jinhui Meng
- School of Life Science, Liaoning University, Shenyang, Liaoning, China
| | - Li Zhang
- School of Life Science, Liaoning University, Shenyang, Liaoning, China
- Liaoning Provincial Key Laboratory of Computational Simulation and Information Processing of Biomacromolecules, Liaoning University, Shenyang, Liaoning, China
- Engineering Laboratory for Molecular Simulation and Designing of Drug Molecules of Liaoning, Liaoning University, Shenyang, Liaoning, China
| | - Zhe He
- School of Life Science, Liaoning University, Shenyang, Liaoning, China
| | - Mengfeng Hu
- School of Life Science, Liaoning University, Shenyang, Liaoning, China
| | - Jinhan Liu
- School of Life Science, Liaoning University, Shenyang, Liaoning, China
| | - Wenzhuo Bao
- School of Life Science, Liaoning University, Shenyang, Liaoning, China
| | - Qifeng Tian
- School of Life Science, Liaoning University, Shenyang, Liaoning, China
| | - Huawei Feng
- School of Pharmacy, Liaoning University, Shenyang, Liaoning, China
| | - Hongsheng Liu
- Liaoning Provincial Key Laboratory of Computational Simulation and Information Processing of Biomacromolecules, Liaoning University, Shenyang, Liaoning, China
- Engineering Laboratory for Molecular Simulation and Designing of Drug Molecules of Liaoning, Liaoning University, Shenyang, Liaoning, China
- School of Pharmacy, Liaoning University, Shenyang, Liaoning, China
| |
Collapse
|
7
|
Liu F, Yang Z, Li J, Wu T, Li X, Zhao L, Wang W, Yu W, Zhang G, Xu Y. Targeting programmed cell death in diabetic kidney disease: from molecular mechanisms to pharmacotherapy. Mol Med 2024; 30:265. [PMID: 39707216 DOI: 10.1186/s10020-024-01020-5] [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: 09/23/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024] Open
Abstract
Diabetic kidney disease (DKD), one of the most prevalent microvascular complications of diabetes, arises from dysregulated glucose and lipid metabolism induced by hyperglycemia, resulting in the deterioration of renal cells such as podocytes and tubular epithelial cells. Programmed cell death (PCD), comprising apoptosis, autophagy, ferroptosis, pyroptosis, and necroptosis, represents a spectrum of cell demise processes intricately governed by genetic mechanisms in vivo. Under physiological conditions, PCD facilitates the turnover of cellular populations and serves as a protective mechanism to eliminate impaired podocytes or tubular epithelial cells, thereby preserving renal tissue homeostasis amidst hyperglycemic stress. However, existing research predominantly elucidates individual modes of cell death, neglecting the intricate interplay and mutual modulation observed among various forms of PCD. In this comprehensive review, we delineate the diverse regulatory mechanisms governing PCD and elucidate the intricate crosstalk dynamics among distinct PCD pathways. Furthermore, we review recent advancements in understanding the pathogenesis of PCD and explore their implications in DKD. Additionally, we explore the potential of natural products derived primarily from botanical sources as therapeutic agents, highlighting their multifaceted effects on modulating PCD crosstalk, thereby proposing novel strategies for DKD treatment.
Collapse
Affiliation(s)
- Fengzhao Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhenyu Yang
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jixin Li
- Xi Yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Tao Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiangyu Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Lijuan Zhao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Wenru Wang
- Xi Yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Wenfei Yu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Guangheng Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yunsheng Xu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250001, China.
| |
Collapse
|
8
|
Tsea I, Olsen TK, Polychronopoulos PA, Tümmler C, Sykes DB, Baryawno N, Dyberg C. DHODH Inhibition Suppresses MYC and Inhibits the Growth of Medulloblastoma in a Novel In Vivo Zebrafish Model. Cancers (Basel) 2024; 16:4162. [PMID: 39766063 PMCID: PMC11674817 DOI: 10.3390/cancers16244162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Medulloblastoma (MB) is the most common high-grade paediatric brain tumour, with group 3 MB patients having the worst prognosis. A high prevalence of group 3 tumours shows overexpression of the MYC oncogene, making it a potential therapeutic target. However, attempts to directly inhibit MYC have so far demonstrated limited success. Dihydroorotate dehydrogenase (DHODH), a crucial enzyme of the pyrimidine biosynthesis process, has emerged as an up-and-coming target in oncology, as its inhibition has shown promise in several cancers. METHODS In this study, we investigated the efficacy of brequinar, a DHODH inhibitor, in MB, with a focus on group 3. In vitro, BRQ's effects on cell viability and MYC expression were tested in seven MB cell lines. In vivo, a novel zebrafish xenograft model was used to evaluate BRQ's impact on tumour growth and toxicity. RESULTS High DHODH expression was identified in group 3 and shh MB subgroups, correlating with poor survival and MYC expression. BRQ demonstrated nanomolar efficacy in inducing apoptosis and reducing MYC expression in group 3 MB cell lines. Finally, we established a novel zebrafish xenograft model and demonstrated that BRQ significantly inhibited tumour growth at non-toxic concentrations in vivo, particularly in the D458 metastatic MB cell line. CONCLUSIONS Our findings indicate that DHODH is a promising therapeutic target in group 3 MBs. Furthermore, BRQ shows potential for clinical application, effectively reducing tumour growth and MYC expression in vitro and in vivo. Moreover, our newly established zebrafish xenograft model offers a promising avenue for rapid in vivo drug testing for use in MB.
Collapse
Affiliation(s)
- Ioanna Tsea
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Thale Kristin Olsen
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Immunology, Genetics, and Pathology, Uppsala University, 753 10 Uppsala, Sweden
| | - Panagiotis Alkinoos Polychronopoulos
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Conny Tümmler
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - David B. Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ninib Baryawno
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Cecilia Dyberg
- Division of Pediatric Oncology and Pediatric Surgery, Department of Women’s and Children’s Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
9
|
Li Y, Li L, Zhang Y, Yun Q, Du R, Ye H, Li Z, Gao Q. Lipocalin-2 silencing alleviates sepsis-induced liver injury through inhibition of ferroptosis. Ann Hepatol 2024; 30:101756. [PMID: 39662594 DOI: 10.1016/j.aohep.2024.101756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/23/2024] [Accepted: 10/23/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION AND OBJECTIVES Liver plays a key role in sepsis, a systemic inflammatory response syndrome caused by infection. Ferroptosis is involved in sepsis-induced liver injury. We aimed to assess the changes in ferroptosis in cecal ligation and puncture (CLP)-induced septic mice, and determine the role of lipocalin-2 (LCN2) in liver ferroptosis. MATERIALS AND METHODS CLP was used to induce sepsis in mice. The morphological changes in liver tissues and mitochondrial structure were observed using hematoxylin and eosin staining and transmission electron microscopy. The levels of serum alanine transaminase, aspartate aminotransferase, superoxide dismutase, and malondialdehyde were detected using the corresponding kits. The changes of reactive oxygen species level in liver tissues were detected using dihydroethidium as a fluorescence probe. LCN2, cysteine-glutamate reverse transport system, and dihydroorotate dehydrogenase protein levels in the liver were detected by western blotting. The ferroptosis inhibitor ferrostatin-1 (Fer-1), iron chelator dexrazoxane (DXZ), iron-dextran, and LCN2 knockdown studies were performed to determine role of ferroptosis and LCN2 in liver injury during sepsis. RESULTS Ferroptosis levels increased in the liver tissues of CLP-induced septic mice. Both Fer-1 and DXZ suppressed ferroptosis and attenuated liver injury following sepsis challenge, whereas iron-dextran increased ferroptosis and liver injury in mice with sepsis. LCN2 knockdown suppressed ferroptosis and reduced oxidative stress in the liver. CONCLUSIONS Ferroptosis inhibition attenuates septic liver injury. LCN2 knockdown alleviates sepsis-induced liver injury by inhibiting ferroptosis and reducing oxidative stress.
Collapse
Affiliation(s)
- Yuping Li
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Anhui Nerve Regeneration Technology and Medical new Materials Engineering Research Center, Bengbu, Anhui 233000, PR China.
| | - Lu Li
- Department of Physiology, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Cardiovascular and cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui 233000, PR China.
| | - Yuming Zhang
- Department of Physiology, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Cardiovascular and cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui 233000, PR China.
| | - Qi Yun
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Cardiovascular and cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui 233000, PR China.
| | - Ruoli Du
- Department of Physiology, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Cardiovascular and cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui 233000, PR China.
| | - Hongwei Ye
- Department of Physiology, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Cardiovascular and cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui 233000, PR China.
| | - Zhenghong Li
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Department of Physiology, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Anhui Nerve Regeneration Technology and Medical new Materials Engineering Research Center, Bengbu, Anhui 233000, PR China.
| | - Qin Gao
- Department of Physiology, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Cardiovascular and cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui 233000, PR China.
| |
Collapse
|
10
|
Guo X, Wang K, Chen H, Wang N, Qiu D, Huang H, Luo J, Xu A, Xu L, Yu Z, Li Y, Zhang H. Leflunomide-Induced Weight Loss: Involvement of DAHPS Activity and Synthesis of Aromatic Amino Acids. Metabolites 2024; 14:645. [PMID: 39590880 PMCID: PMC11596867 DOI: 10.3390/metabo14110645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Leflunomide, an isoxazole immunosuppressant, is widely used in the treatment of diseases such as rheumatoid arthritis (RA) and psoriatic arthritis (PsA) as well as lupus nephritis (LN). In recent years, clinical data have shown that some patients have obvious weight loss, liver injury, and other serious adverse reactions after taking leflunomide. However, the causes and mechanisms by which leflunomide reduces weight are unclear. Methods: Therefore, we used a mouse animal model to administer leflunomide, and we observed that the weight of mice in the leflunomide experimental group was significantly reduced (p < 0.01). In this animal experiment, a metabolomic method was used to analyze the livers of the mice in the experimental group and found that the main difference in terms of metabolic pathways was in the metabolism of aromatic amino acids, and it was confirmed that leflunomide can inhibit the limitations of phenylalanine, tyrosine, and tryptophan biosynthesis. Results: Our study revealed that leflunomide inhibited the activity of DAHPS in the gut microbiota, disrupting the metabolism of phenylalanine, tyrosine, and tryptophan, as well as the metabolism of carbohydrates and lipids. Leflunomide also increased endoplasmic reticulum stress by activating the PERK pathway, thereby promoting CHOP expression and increasing apoptosis-induced liver damage. Conclusions: These effects may be related to the observed weight loss induced by leflunomide.
Collapse
Affiliation(s)
- Xiaoyu Guo
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China; (X.G.); (K.W.); (H.C.); (N.W.); (L.X.); (Z.Y.)
| | - Kai Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China; (X.G.); (K.W.); (H.C.); (N.W.); (L.X.); (Z.Y.)
| | - Hongli Chen
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China; (X.G.); (K.W.); (H.C.); (N.W.); (L.X.); (Z.Y.)
| | - Na Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China; (X.G.); (K.W.); (H.C.); (N.W.); (L.X.); (Z.Y.)
| | - Dongmei Qiu
- School of Medicine and Health, Wuhan Polytechnic University, Wuhan 430023, China; (D.Q.); (H.H.); (J.L.); (A.X.)
| | - Haiyun Huang
- School of Medicine and Health, Wuhan Polytechnic University, Wuhan 430023, China; (D.Q.); (H.H.); (J.L.); (A.X.)
| | - Jiyu Luo
- School of Medicine and Health, Wuhan Polytechnic University, Wuhan 430023, China; (D.Q.); (H.H.); (J.L.); (A.X.)
| | - Ao Xu
- School of Medicine and Health, Wuhan Polytechnic University, Wuhan 430023, China; (D.Q.); (H.H.); (J.L.); (A.X.)
| | - Lingyun Xu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China; (X.G.); (K.W.); (H.C.); (N.W.); (L.X.); (Z.Y.)
| | - Zejun Yu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China; (X.G.); (K.W.); (H.C.); (N.W.); (L.X.); (Z.Y.)
| | - Yuanyuan Li
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongling Zhang
- School of Medicine and Health, Wuhan Polytechnic University, Wuhan 430023, China; (D.Q.); (H.H.); (J.L.); (A.X.)
| |
Collapse
|
11
|
Shahhiran MAA, Abdul Kadir MF, Nor Rashid N, Abdul-Rahman PS, Othman S. Mechanisms of S-phase arrest and mitochondrial dysfunction in complex III by DHODH inhibitors in tumorigenic TNBC cells. Histochem Cell Biol 2024; 163:3. [PMID: 39557682 DOI: 10.1007/s00418-024-02339-0] [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] [Accepted: 09/26/2024] [Indexed: 11/20/2024]
Abstract
Dihydroorotate dehydrogenase (DHODH) inhibitors have recently gained increasing research interest owing to their potential for treating breast cancers. We explored their effects in different breast cancer subtypes, focusing on mitochondrial dysfunction. The sensitivity of different subtypes to the inhibitors was investigated with respect to DHODH expression, tumorigenic, and receptor status. Analysis of respiratory complexes, cell cycle, reactive oxygen species (ROS), and cell differentiation were performed. Four cell lines with different receptor status were included, namely MCF-7, MDAMB-231, SKBR-3, and MCF-10A. We showed that MCF-7 and MDAMB-231 cells of the subtypes (ER+/PR+/HER2-) and (ER-/PR-/HER2-), respectively, were responsive to brequinar. Brequinar (BQR) caused cell cycle arrest in the S-phase in sensitive subtypes of breast cells but induced cell differentiation only in poorly differentiated breast cells. All cell subtypes showed increased generation of ROS, both intracellular and mitochondrial ROS with a greater increase seen in mitochondrial ROS in response to DHODH inhibitor, subsequently contributing to mitochondrial dysfunction. BQR also disrupts the function of complex III in ER+/PR+ and triple negative breast cancer (TNBC) subtypes. Collectively, we have found that MDAMB-231 TNBC cell was the most affected by DHODH inhibition in terms of sensitivity, cell cycle arrest, induction of cell differentiation, production of ROS, and mitochondrial complexes disruption. In conclusion, these findings suggest that DHODH inhibitors can potentially become a valuable targeted therapy for TNBC subtype and further consolidates its therapeutic potential as part of the combinatorial therapy against this resilient breast cancer subtype.
Collapse
Affiliation(s)
- Muhammad Aiman Akmal Shahhiran
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Drug Design and Development Research Group, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mohamad Fairus Abdul Kadir
- Aurigene Discovery Technology (M) Sdn Bhd, Level 2 Research Management and Innovation Complex, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nurshamimi Nor Rashid
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
- Drug Design and Development Research Group, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | | | - Shatrah Othman
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
- Drug Design and Development Research Group, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
12
|
DeRatt LG, Zhang Z, Pietsch EC, Cisar J, Wang A, Wang CY, Tanner A, Shaffer P, Jacoby E, Kazmi F, Shukla N, Philippar U, Attar RM, Edwards JP, Kuduk SD. Identification of isoquinolinone DHODH inhibitor isosteres. Bioorg Med Chem Lett 2024; 113:129965. [PMID: 39284456 DOI: 10.1016/j.bmcl.2024.129965] [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: 07/12/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 10/07/2024]
Abstract
DHODH inhibition represents an attractive approach to overcome differentiation blockade for the treatment of AML. In a previous communication, we described our efforts leading to the discovery of compound 3 (JNJ-74856665), an orally bioavailable, potent, and selective DHODH inhibitor for clinical development. Guided by the co-crystal structures bound to human DHODH, other fused six-membered constructs were explored as isosteric replacements of the isoquinolinone central core. The correct positioning of the nitrogen in these core systems proved to be essential in modulating potency. Herein is described the synthesis of these complexly functionalized cores and their profiling, leading to DHODH inhibitors that possess favorable properties suitable for further development.
Collapse
Affiliation(s)
| | - Zhuming Zhang
- Janssen Research and Development, Spring House, PA 19477, USA
| | | | - Justin Cisar
- Janssen Research and Development, Spring House, PA 19477, USA
| | - Aihua Wang
- Janssen Research and Development, Spring House, PA 19477, USA
| | - Chao-Yuan Wang
- Janssen Research and Development, Spring House, PA 19477, USA
| | | | - Paul Shaffer
- Janssen Research and Development, Spring House, PA 19477, USA
| | - Edgar Jacoby
- Janssen Research and Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Faraz Kazmi
- Janssen Research and Development, Spring House, PA 19477, USA
| | - Neetu Shukla
- Janssen Research and Development, Spring House, PA 19477, USA
| | - Ulrike Philippar
- Janssen Research and Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Ricardo M Attar
- Janssen Research and Development, Spring House, PA 19477, USA
| | - James P Edwards
- Janssen Research and Development, Spring House, PA 19477, USA
| | - Scott D Kuduk
- Janssen Research and Development, Spring House, PA 19477, USA.
| |
Collapse
|
13
|
Herrmann A, Gege C, Wangen C, Wagner S, Kögler M, Cordsmeier A, Irrgang P, Ip WH, Weil T, Hunszinger V, Groß R, Heinen N, Pfaender S, Reuter S, Klopfleisch R, Uhlig N, Eberlein V, Issmail L, Grunwald T, Hietel B, Cynis H, Münch J, Sparrer KMJ, Ensser A, Tenbusch M, Dobner T, Vitt D, Kohlhof H, Hahn F. Orally bioavailable RORγ/DHODH dual host-targeting small molecules with broad-spectrum antiviral activity. Antiviral Res 2024; 231:106008. [PMID: 39306285 DOI: 10.1016/j.antiviral.2024.106008] [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: 07/19/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
Host-directed antivirals (HDAs) represent an attractive treatment option and a strategy for pandemic preparedness, especially due to their potential broad-spectrum antiviral activity and high barrier to resistance development. Particularly, dual-targeting HDAs offer a promising approach for antiviral therapy by simultaneously disrupting multiple pathways essential for viral replication. Izumerogant (IMU-935) targets two host proteins, (i) the retinoic acid receptor-related orphan receptor γ isoform 1 (RORγ1), which modulates cellular cholesterol metabolism, and (ii) the enzyme dihydroorotate dehydrogenase (DHODH), which is involved in de novo pyrimidine synthesis. Here, we synthesized optimized derivatives of izumerogant and characterized their antiviral activity in comparison to a recently described structurally distinct RORγ/DHODH dual inhibitor. Cell culture-based infection models for enveloped and non-enveloped DNA and RNA viruses, as well as a retrovirus, demonstrated high potency and broad-spectrum activity against human viral pathogens for RORγ/DHODH dual inhibitors at nanomolar concentrations. Comparative analyses with equipotent single-target inhibitors in metabolite supplementation approaches revealed that the dual-targeting mode represents the mechanistic basis for the potent antiviral activity. For SARS-CoV-2, an optimized dual inhibitor completely blocked viral replication in human airway epithelial cells at 5 nM and displayed a synergistic drug interaction with the nucleoside analog molnupiravir. In a SARS-CoV-2 mouse model, treatment with a dual inhibitor alone, or in combination with molnupiravir, reduced the viral load by 7- and 58-fold, respectively. Considering the clinical safety, oral bioavailability, and tolerability of izumerogant in a recent Phase I study, izumerogant-like drugs represent potent dual-targeting antiviral HDAs with pronounced broad-spectrum activity for further clinical development.
Collapse
Affiliation(s)
| | | | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Sabrina Wagner
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Melanie Kögler
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Arne Cordsmeier
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Pascal Irrgang
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Wing-Hang Ip
- Leibniz Institute of Virology, Hamburg, Germany.
| | - Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Victoria Hunszinger
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Natalie Heinen
- Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, Germany.
| | - Stephanie Pfaender
- Leibniz Institute of Virology, Hamburg, Germany; Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, Germany; University of Luebeck, Department of Natural Sciences, Institute of Virology and Cell Biology, Lübeck, Germany.
| | - Sebastian Reuter
- University Hospital Essen - Ruhrlandklinik, Department of Pulmonary Medicine, Experimental Pneumology, Essen, Germany.
| | - Robert Klopfleisch
- Institute for Animal Pathology, Freie Universität Berlin, Berlin, Germany.
| | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Benjamin Hietel
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Halle, Germany.
| | - Holger Cynis
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Halle, Germany; Junior Research Group "Immunomodulation in Pathophysiological Processes", Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany.
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | | | - Armin Ensser
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Matthias Tenbusch
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | | | | | | | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
14
|
Yang L, Ma D, Liu S, Zou L. The DHODH inhibitor teriflunomide impedes cell proliferation and enhances chemosensitivity to daunorubicin (DNR) in T-cell acute lymphoblastic leukemia. Ann Hematol 2024:10.1007/s00277-024-05998-0. [PMID: 39377943 DOI: 10.1007/s00277-024-05998-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological tumor that requires novel treatment strategies, especially for relapsed/refractory cases. Dihydroorotate dehydrogenase (DHODH), a key enzyme in the de novo pyrimidine synthesis pathway, has been identified as a potential target for tumors. Besides, Teriflunomide (TRF) is a DHODH inhibitor with anticancer effects; however, its role in T-ALL remains poorly understood. Here, we investigated the potential anticancer effects of TRF on T-ALL cells, and the results showed that TRF inhibited cell proliferation, caused S-phase cell cycle arrest, and promoted apoptosis of T-ALL (MOLT4 and JURKAT) cell lines. In addition, TRF reduced the infiltration capacity of T-ALL cells in T-ALL xenograft mice while up-regulating the expression of P53 and BTG2. The BTG2 knockdown significantly attenuated the inhibitory effect of TRF on cellular growth and suppressed the TRF-mediated elevated expression of P53 in T-ALL cells. Moreover, combined treatment with TRF and daunorubicin (DNR) significantly reduced cell viability and promoted apoptosis in DNR-resistant T-ALL cells. Our study provides valuable insights into the critical role of TRF in treating T-ALL while increasing the sensitivity of DNR-resistant T-ALL cells to DNR.
Collapse
Affiliation(s)
- Li Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Center for Clinical Molecular Laboratory Medicine of Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Deyu Ma
- Center for Clinical Molecular Laboratory Medicine of Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders (Chongqing), Chongqing, 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Shan Liu
- Center for Clinical Molecular Laboratory Medicine of Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- National Clinical Research Center for Child Health and Disorders (Chongqing), Chongqing, 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Lin Zou
- Center for Clinical Molecular Laboratory Medicine of Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Clinical Research Unit, Children's Hospital of Shanghai Jiao Tong University, 355 Luding Rd, Putuo District, Shanghai, 200062, China.
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, China.
| |
Collapse
|
15
|
Wang J, Wang X, Yang C, Li Q, Li D, Du X, Cheng Y, Tian M, Zheng L, Tong Q. circE2F1-encoded peptide inhibits circadian machinery essential for nucleotide biosynthesis and tumor progression via repressing SPIB/E2F1 axis. Int J Biol Macromol 2024; 280:135698. [PMID: 39288851 DOI: 10.1016/j.ijbiomac.2024.135698] [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: 05/21/2024] [Revised: 09/01/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Circadian clock dominates a variety of biological activities, while its roles and regulatory mechanisms in neuroblastoma (NB), a pediatric extracranial malignancy, still remain largely elusive. Herein, through comprehensive analyses of public datasets, E2F transcription factor 1 (E2F1) and its circular RNA (circE2F1)-encoded 99-amino acid peptide (E2F1-99aa) were identified as vital regulators of circadian machinery essential for purine and pyrimidine biosynthesis during NB progression. Mechanistically, through interaction with Spi-B transcription factor (SPIB), E2F1 was transactivated to up-regulate circadian machinery genes (CRY1 and TIMELESS), resulting in relief of CLOCK/BMAL1-repressed transcription of enzymes (DHODH, PAICS, or PPAT) essential for de novo purine and pyrimidine biosynthesis. The biogenesis of circE2F1 was repressed by eukaryotic translation initiation factor 4A3 (EIF4A3), while E2F1-99aa or its truncated peptide competitively bound to SPIB, leading to decrease in SPIB-E2F1 interaction, circadian machinery and nucleotide biosynthetic gene expression, purine or pyrimidine biosynthesis, tumorigenesis, and aggresiveness of NB cells. In clinical NB cases, high EIF4A3, E2F1 or SPIB expression was correlated with low survival possibility of patients, while lower circE2F1 or E2F1-99aa levels were associated with advanced stages and tumor progression. These results indicate that circE2F1-encoded peptide inhibits circadian machinery essential for nucleotide biosynthesis and tumor progression via repressing SPIB/E2F1 axis.
Collapse
Affiliation(s)
- Jianqun Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Xiaojing Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China; Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Chunhui Yang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Qilan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Xinyi Du
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Yang Cheng
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Minxiu Tian
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China.
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China.
| |
Collapse
|
16
|
Zhang JF, Hong LH, Fan SY, Zhu L, Yu ZP, Chen C, Kong LY, Luo JG. Discovery of piperine derivatives as inhibitors of human dihydroorotate dehydrogenase to induce ferroptosis in cancer cells. Bioorg Chem 2024; 150:107594. [PMID: 38941701 DOI: 10.1016/j.bioorg.2024.107594] [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: 05/18/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
Inhibition of human dihydroorotate dehydrogenase (hDHODH) represents a promising strategy for suppressing the proliferation of cancer cells. To identify novel and potent hDHODH inhibitors, a total of 28 piperine derivatives were designed and synthesized. Their cytotoxicities against three human cancer cell lines (NCI-H226, HCT-116, and MDA-MB-231) and hDHODH inhibitory activities were also evaluated. Among them, compound H19, exhibited the strongest inhibitory activities (NCI-H226 IC50 = 0.95 µM, hDHODH IC50 = 0.21 µM). Further pharmacological investigations revealed that H19 exerted anticancer effects by inducing ferroptosis in NCI-H226 cells, with its cytotoxicity being reversed by ferroptosis inhibitors. This was supported by the intracellular growth or decline of ferroptosis markers, including lipid peroxidation, Fe2+, GSH, and 4-HNE. Overall, H19 emerges as a promising hDHODH inhibitor with potential anticancer properties warranting development.
Collapse
Affiliation(s)
- Jian-Fei Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Li-Hong Hong
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Shi-Ying Fan
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Ling Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Zhan-Peng Yu
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Chen Chen
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Jian-Guang Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research, State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
17
|
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
|
18
|
DeRatt LG, Zhang Z, Pietsch C, Cisar JS, Zhang X, Wang W, Tanner A, Matico R, Shaffer P, Jacoby E, Kazmi F, Shukla N, Bush TL, Patrick A, Philippar U, Attar R, Edwards JP, Kuduk SD. Discovery of JNJ-74856665: A Novel Isoquinolinone DHODH Inhibitor for the Treatment of AML. J Med Chem 2024; 67:11254-11272. [PMID: 38889244 DOI: 10.1021/acs.jmedchem.4c00809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Acute myelogenous leukemia (AML), a heterogeneous disease of the blood and bone marrow, is characterized by the inability of myeloblasts to differentiate into mature cell types. Dihydroorotate dehydrogenase (DHODH) is an enzyme well-known in the pyrimidine biosynthesis pathway and preclinical findings demonstrated that DHODH is a metabolic vulnerability in AML as inhibitors can induce differentiation across multiple AML subtypes. As a result of virtual screening and structure-based drug design approaches, a novel series of isoquinolinone DHODH inhibitors was identified. Further lead optimization afforded JNJ-74856665 as an orally bioavailable, potent, and selective DHODH inhibitor with favorable physicochemical properties selected for clinical development in patients with AML and myelodysplastic syndromes (MDS).
Collapse
Affiliation(s)
- Lindsey G DeRatt
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Zhuming Zhang
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Christine Pietsch
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Justin S Cisar
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Xiaochun Zhang
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Weixue Wang
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Alexandra Tanner
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Rosalie Matico
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Paul Shaffer
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Edgar Jacoby
- Janssen Research and Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Faraz Kazmi
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Neetu Shukla
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Tammy L Bush
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Aaron Patrick
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Ulrike Philippar
- Janssen Research and Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Ricardo Attar
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - James P Edwards
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| | - Scott D Kuduk
- Janssen Research and Development, Spring House, Pennsylvania 19477, United States
| |
Collapse
|
19
|
Dong S, Zhang M, Cheng Z, Zhang X, Liang W, Li S, Li L, Xu Q, Song S, Liu Z, Yang G, Zhao X, Tao Z, Liang S, Wang K, Zhang G, Hu S. Redistribution of defective mitochondria-mediated dihydroorotate dehydrogenase imparts 5-fluorouracil resistance in colorectal cancer. Redox Biol 2024; 73:103207. [PMID: 38805974 PMCID: PMC11152977 DOI: 10.1016/j.redox.2024.103207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
Although 5-fluorouracil (5-FU) is the primary chemotherapy treatment for colorectal cancer (CRC), its efficacy is limited by drug resistance. Ferroptosis activation is a promising treatment for 5-FU-resistant cancer cells; however, potential therapeutic targets remain elusive. This study investigated ferroptosis vulnerability and dihydroorotate dehydrogenase (DHODH) activity using stable, 5-FU-resistant CRC cell lines and xenograft models. Ferroptosis was characterized by measuring malondialdehyde levels, assessing lipid metabolism and peroxidation, and using mitochondrial imaging and assays. DHODH function is investigated through gene knockdown experiments, tumor behavior assays, mitochondrial import reactions, intramitochondrial localization, enzymatic activity analyses, and metabolomics assessments. Intracellular lipid accumulation and mitochondrial DHODH deficiency led to lipid peroxidation overload, weakening the defense system of 5-FU-resistant CRC cells against ferroptosis. DHODH, primarily located within the inner mitochondrial membrane, played a crucial role in driving intracellular pyrimidine biosynthesis and was redistributed to the cytosol in 5-FU-resistant CRC cells. Cytosolic DHODH, like its mitochondrial counterpart, exhibited dihydroorotate catalytic activity and participated in pyrimidine biosynthesis. This amplified intracellular pyrimidine pools, thereby impeding the efficacy of 5-FU treatment through molecular competition. These findings contribute to the understanding of 5-FU resistance mechanisms and suggest that ferroptosis and DHODH are promising therapeutic targets for patients with CRC exhibiting resistance to 5-FU.
Collapse
Affiliation(s)
- Shuohui Dong
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China
| | - Mingguang Zhang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiqiang Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China
| | - Weili Liang
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China
| | - Songhan Li
- Department of General Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Linchuan Li
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Qian Xu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Siyi Song
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Zitian Liu
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China
| | - Guangwei Yang
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China
| | - Xiang Zhao
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China
| | - Ze Tao
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China
| | - Shuo Liang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, No. 4, Duanxing West Road, Jinan, Shandong,250022, China.
| | - Kexin Wang
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China.
| | - Guangyong Zhang
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, 250014, China.
| | - Sanyuan Hu
- Department of General Surgery, Qilu Hospital of Shandong University, No. 107 Wenhua Xilu, Jinan, Shandong, 250012, China.
| |
Collapse
|
20
|
Ishikawa C, Mori N. Pivotal role of dihydroorotate dehydrogenase as a therapeutic target in adult T-cell leukemia. Eur J Haematol 2024; 113:99-109. [PMID: 38558052 DOI: 10.1111/ejh.14209] [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: 11/17/2023] [Revised: 03/10/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES We aimed to determine the role of dihydroorotate dehydrogenase (DHODH) in pathogenesis of adult T-cell leukemia (ATL) caused by human T-cell leukemia virus type 1 (HTLV-1) and the effects of its inhibition on the de novo pyrimidine biosynthesis pathway. METHODS Cell proliferation, viability, cycle, and apoptosis were analyzed using WST-8 assays, flow cytometry, and Hoechst 33342 staining. To elucidate the molecular mechanisms involved in the anti-ATL effects of DHODH knockdown and inhibition, RT-PCR and immunoblotting were conducted. RESULTS HTLV-1-infected T-cell lines aberrantly expressed DHODH. Viral infection and the oncoprotein, Tax, enhanced DHODH expression, while knockdown of DHODH decreased HTLV-1-infected T-cell growth. In addition, BAY2402234, a DHODH inhibitor, exerted an anti-proliferative effect, which was reversed by uridine supplementation. BAY2402234 induced DNA damage and S phase arrest by downregulating c-Myc, CDK2, and cyclin A and upregulating p53 and cyclin E. It also induced caspase-mediated apoptosis by the upregulation of pro-apoptotic and downregulation of anti-apoptotic proteins. Furthermore, BAY2402234 induced caspase-independent ferroptosis and necroptosis. It decreased phosphorylation of IKK, IκBα, PTEN, Akt, and its downstream targets, suggesting that inhibition of NF-κB and Akt signaling is involved in its anti-ATL action. CONCLUSION These findings highlight DHODH as a potential therapeutic target for treating ATL.
Collapse
Affiliation(s)
- Chie Ishikawa
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Division of Health Sciences, Transdisciplinary Research Organization for Subtropics and Island Studies, University of the Ryukyus, Nishihara, Japan
| | - Naoki Mori
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| |
Collapse
|
21
|
Liu L, Mo W, Chen M, Qu Y, Wang P, Liang Y, Yan X. Targeted inhibition of DHODH is synergistic with BCL2 blockade in HGBCL with concurrent MYC and BCL2 rearrangement. BMC Cancer 2024; 24:761. [PMID: 38918775 PMCID: PMC11197201 DOI: 10.1186/s12885-024-12534-w] [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: 10/27/2023] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
High-grade B-cell lymphoma (HGBCL), the subtype of non-Hodgkin lymphoma, to be relapsed or refractory in patients after initial therapy or salvage chemotherapy. Dual dysregulation of MYC and BCL2 is one of the important pathogenic mechanisms. Thus, combined targeting of MYC and BCL2 appears to be a promising strategy. Dihydroorotate dehydrogenase (DHODH) is the fourth rate-limiting enzyme for the de novo biosynthesis of pyrimidine. It has been shown to be a potential therapeutic target for multiple diseases. In this study, the DHODH inhibitor brequinar exhibited growth inhibition, cell cycle blockade, and apoptosis promotion in HGBCL cell lines with MYC and BCL2 rearrangements. The combination of brequinar and BCL2 inhibitors venetoclax had a synergistic inhibitory effect on the survival of DHL cells through different pathways. Venetoclax could upregulate MCL-1 and MYC expression, which has been reported as a resistance mechanism of BCL2 inhibitors. Brequinar downregulated MCL-1 and MYC, which could potentially overcome drug resistance to venetoclax in HGBCL cells. Furthermore, brequinar could downregulate a broad range of genes, including ribosome biosynthesis genes, which might contribute to its anti-tumor effects. In vivo studies demonstrated synergetic tumor growth inhibition in xenograft models with brequinar and venetoclax combination treatment. These results provide preliminary evidence for the rational combination of DHODH and BCL2 blockade in HGBCL with abnormal MYC and BCL2.
Collapse
Affiliation(s)
- Lin Liu
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Wenbin Mo
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Miao Chen
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yi Qu
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Pingping Wang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Ying Liang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaojing Yan
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
- , No. 155, North Nanjing Road, Heping District, Shenyang, 110001, China.
| |
Collapse
|
22
|
Zhao BQ, Chen J, Chen JX, Cheng Y, Zhou JF, Bai JS, Mao DY, Zhou B. Classical swine fever virus non-structural protein 4A recruits dihydroorotate dehydrogenase to facilitate viral replication. J Virol 2024; 98:e0049424. [PMID: 38757985 PMCID: PMC11237749 DOI: 10.1128/jvi.00494-24] [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: 03/15/2024] [Accepted: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
Mitochondria are energy producers in cells, which can affect viral replication by regulating the host innate immune signaling pathways, and the changes in their biological functions are inextricably linked the viral life cycle. In this study, we screened a library of 382 mitochondria-targeted compounds and identified the antiviral inhibitors of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme in the de novo synthesis pathway of pyrimidine ribonucleotides, against classical swine fever virus (CSFV). Our data showed that the inhibitors interfered with viral RNA synthesis in a dose-dependent manner, with half-maximal effective concentrations (EC50) ranging from 0.975 to 26.635 nM. Remarkably, DHODH inhibitors obstructed CSFV replication by enhancing the innate immune response including the TBK1-IRF3-STAT1 and NF-κB signaling pathways. Furthermore, the data from a series of compound addition and supplementation trials indicated that DHODH inhibitors also inhibited CSFV replication by blocking the de novo pyrimidine synthesis. Remarkably, DHODH knockdown demonstrated that it was essential for CSFV replication. Mechanistically, confocal microscopy and immunoprecipitation assays showed that the non-structural protein 4A (NS4A) recruited and interacted with DHODH in the perinuclear. Notably, NS4A enhanced the DHODH activity and promoted the generation of UMP for efficient viral replication. Structurally, the amino acids 65-229 of DHODH and the amino acids 25-40 of NS4A were pivotal for this interaction. Taken together, our findings highlight the critical role of DHODH in the CSFV life cycle and offer a potential antiviral target for the development of novel therapeutics against CSF. IMPORTANCE Classical swine fever remains one of the most economically important viral diseases of domestic pigs and wild boar worldwide. dihydroorotate dehydrogenase (DHODH) inhibitors have been shown to suppress the replication of several viruses in vitro and in vivo, but the effects on Pestivirus remain unknown. In this study, three specific DHODH inhibitors, including DHODH-IN-16, BAY-2402234, and Brequinar were found to strongly suppress classical swine fever virus (CSFV) replication. These inhibitors target the host DHODH, depleting the pyrimidine nucleotide pool to exert their antiviral effects. Intriguingly, we observed that the non-structural protein 4A of CSFV induced DHODH to accumulate around the nucleus in conjunction with mitochondria. Moreover, NS4A exhibited a strong interaction with DHODH, enhancing its activity to promote efficient CSFV replication. In conclusion, our findings enhance the understanding of the pyrimidine synthesis in CSFV infection and expand the novel functions of CSFV NS4A in viral replication, providing a reference for further exploration of antiviral targets against CSFV.
Collapse
Affiliation(s)
- Bing-qian Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jing Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jin-Xia Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yan Cheng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiang-fei Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ji-shan Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ding-yi Mao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Bin Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
23
|
Baiskhanova D, Schäfer H. The Role of Nrf2 in the Regulation of Mitochondrial Function and Ferroptosis in Pancreatic Cancer. Antioxidants (Basel) 2024; 13:696. [PMID: 38929135 PMCID: PMC11201043 DOI: 10.3390/antiox13060696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) represents the master regulator of the cellular antioxidant response and plays a critical role in tumorigenesis. This includes a preventive effect of Nrf2 on cell death through ferroptosis, which represents an essential mechanism of therapy resistance in malignant tumors, such as pancreatic ductal adenocarcinoma (PDAC) as one of the most aggressive and still incurable tumors. Addressing this issue, we provide an overview on Nrf2 mediated antioxidant response with particular emphasis on its effect on mitochondria as the organelle responsible for the execution of ferroptosis. We further outline how deregulated Nrf2 adds to the progression and therapy resistance of PDAC, especially with respect to the role of ferroptosis in anti-cancer drug mediated cell killing and how this is impaired by Nrf2 as an essential mechanism of drug resistance. Our review further discusses recent approaches for Nrf2 inhibition by natural and synthetic compounds to overcome drug resistance based on enhanced ferroptosis. Finally, we provide an outlook on therapeutic strategies based on Nrf2 inhibition combined with ferroptosis inducing drugs.
Collapse
Affiliation(s)
- Dinara Baiskhanova
- Laboratory of Molecular Gastroenterology and Tumor Biology, Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany;
| | | |
Collapse
|
24
|
Ma Y, Cong L, Shen W, Yang C, Ye K. Ferroptosis defense mechanisms: The future and hope for treating osteosarcoma. Cell Biochem Funct 2024; 42:e4080. [PMID: 38924104 DOI: 10.1002/cbf.4080] [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: 04/25/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024]
Abstract
Currently, challenges such as chemotherapy resistance, resulting from preoperative and postoperative chemotherapy, postoperative recurrence, and poor bone regeneration quality, are becoming increasingly prominent in osteosarcoma (OS) treatment. There is an urgent need to find more effective ways to address these issues. Ferroptosis is a novel form of iron-dependent programmed cell death, distinct from other forms of cell death. In this paper, we summarize how, through the three major defense systems of ferroptosis, not only can substances from traditional Chinese medicine, antitumor drugs, and nano-drug carriers induce ferroptosis in OS cells, but they can also be combined with immunotherapy, differentiation therapy, and other treatment modalities to significantly enhance chemotherapy sensitivity and inhibit tumor growth. Thus, ferroptosis holds great potential in treating OS, offering more choices and possibilities for future clinical interventions.
Collapse
Affiliation(s)
- Yulong Ma
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Liming Cong
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Wenxiang Shen
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Chunwang Yang
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Kaishan Ye
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
25
|
Dimitrijević MG, Roschger C, Lang K, Zierer A, Paunović MG, Obradović AD, Matić MM, Pocrnić M, Galić N, Ćirić A, Joksović MD. Discovery of a new class of potent pyrrolo[3,4-c]quinoline-1,3-diones based inhibitors of human dihydroorotate dehydrogenase: Synthesis, pharmacological and toxicological evaluation. Bioorg Chem 2024; 147:107359. [PMID: 38613925 DOI: 10.1016/j.bioorg.2024.107359] [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: 01/18/2024] [Revised: 02/28/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
Twenty N-substituted pyrrolo[3,4-c]quinoline-1,3-diones 3a-t were synthesized by a cyclization reaction of Pfitzinger's quinoline ester precursor with the selected aromatic, heteroaromatic and aliphatic amines. The structures of all derivatives were confirmed by IR, 1H NMR, 13C NMR and HRMS spectra, while their purity was determined using HPLC techniques. Almost all compounds were identified as a new class ofpotent inhibitors against hDHODH among which 3a and 3t were the most active ones with the same IC50 values of 0.11 μM, about seven times better than reference drug leflunomide. These two derivatives also exhibited very low cytotoxic effects toward healthy HaCaT cells and the optimal lipophilic properties with logP value of 1.12 and 2.07 respectively, obtained experimentally at physiological pH. We further evaluated the comparative differences in toxicological impact of the three most active compounds 3a, 3n and 3t and reference drug leflunomide. The rats were divided into five groups and were treated intraperitoneally, control group (group I) with a single dose of leflunomide (20 mg/kg) group II and the other three groups, III, IV and V were treated with 3a, 3n and 3t (20 mg/kg bw) separately. The investigation was performed in liver, kidney and blood by examining serum biochemical parameters and parameters of oxidative stress.
Collapse
Affiliation(s)
- Marina G Dimitrijević
- University of Kragujevac, Faculty of Sciences, Department of Chemistry, R. Domanovića 12, 34000 Kragujevac, Serbia
| | - Cornelia Roschger
- University Clinic for Cardiac-, Vascular- and Thoracic Surgery, Medical Faculty, Johannes Kepler University Linz, Krankenhausstraße 7a, 4020 Linz, Austria
| | - Kevin Lang
- University Clinic for Cardiac-, Vascular- and Thoracic Surgery, Medical Faculty, Johannes Kepler University Linz, Krankenhausstraße 7a, 4020 Linz, Austria
| | - Andreas Zierer
- University Clinic for Cardiac-, Vascular- and Thoracic Surgery, Medical Faculty, Johannes Kepler University Linz, Krankenhausstraße 7a, 4020 Linz, Austria
| | - Milica G Paunović
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, P.O. Box 60, Kragujevac 34000, Serbia
| | - Ana D Obradović
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, P.O. Box 60, Kragujevac 34000, Serbia
| | - Miloš M Matić
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, P.O. Box 60, Kragujevac 34000, Serbia
| | - Marijana Pocrnić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000 Zagreb, Croatia
| | - Nives Galić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000 Zagreb, Croatia
| | - Andrija Ćirić
- University of Kragujevac, Faculty of Sciences, Department of Chemistry, R. Domanovića 12, 34000 Kragujevac, Serbia
| | - Milan D Joksović
- University of Kragujevac, Faculty of Sciences, Department of Chemistry, R. Domanovića 12, 34000 Kragujevac, Serbia.
| |
Collapse
|
26
|
Wang S, Guo Q, Zhou L, Xia X. Ferroptosis: A double-edged sword. Cell Death Discov 2024; 10:265. [PMID: 38816377 PMCID: PMC11139933 DOI: 10.1038/s41420-024-02037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Ferroptosis represents a form of programmed cell death that is propelled by iron-dependent lipid peroxidation, thereby being distinguished by the prominent features of iron accumulation and lipid peroxidation. Ferroptosis has been implicated in numerous physiological and pathological phenomena, with mounting indications that it holds significant implications for cancer and other medical conditions. On one side, it demonstrates anti-cancer properties by triggering ferroptosis within malignant cells, and on the other hand, it damages normal cells causing other diseases. Therefore, in this paper, we propose to review the paradoxical regulation of ferroptosis in tumors and other diseases. First, we introduce the development history, concept and mechanism of ferroptosis. The second part focuses on the methods of inducing ferroptosis in tumors. The third section emphasizes the utilization of ferroptosis in different medical conditions and strategies to inhibit ferroptosis. The fourth part elucidates the key contradictions in the control of ferroptosis. Finally, potential research avenues in associated domains are suggested.
Collapse
Affiliation(s)
- Shengmei Wang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
27
|
Wang S, Li Y, Lin Y, Li J, Guo L, Wang H, Lin X, Liu Z, Zhang B, Liao Z, Zhang Z. Bioinformatics analysis and experimental verification of the cancer-promoting effect of DHODH in clear cell renal cell carcinoma. Sci Rep 2024; 14:11985. [PMID: 38796629 PMCID: PMC11127953 DOI: 10.1038/s41598-024-62738-0] [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: 03/25/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a malignant tumor of the urinary system. To explore the potential mechanisms of DHODH in ccRCC, we analyzed its molecular characteristics using public databases. TCGA pan-cancer dataset was used to analyze DHODH expression in different cancer types and TCGA ccRCC dataset was used to assess differential expression, prognosis correlation, immune infiltration, single-gene, and functional enrichment due to DHODH. The GSCALite and CellMiner databases were employed to explore drugs and perform molecular docking analysis with DHODH. Protein-protein interaction networks and ceRNA regulatory networks of DHODH were constructed using multiple databases. The effect of DHODH on ccRCC was confirmed in vitro. DHODH was highly expressed in ccRCC. Immune infiltration analysis revealed that DHODH may be involved in regulating the infiltration of immunosuppressive cells such as Tregs. Notably, DHODH influenced ccRCC progression by forming regulatory networks with molecules, such as hsa-miR-26b-5p and UMPS and significantly enhanced the malignant characteristics of ccRCC cells. Several drugs, such as lapatinib, silmitasertib, itraconazole, and dasatinib, were sensitive to DHODH expression and exhibited strong molecular binding with it. Thus, DHODH may promote ccRCC progression and is a candidate effective therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Songsong Wang
- Department of Urology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
- School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yan Li
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Yilong Lin
- School of Medicine, Xiamen University, Xiamen, 361000, China
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Junting Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Lang Guo
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Haoyu Wang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Xinyuan Lin
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Ziming Liu
- Department of Urology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.
- School of Medicine, Xiamen University, Xiamen, 361000, China.
| | - Bingqi Zhang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Zhengming Liao
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Zhongmin Zhang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| |
Collapse
|
28
|
Xie T, Qin C, Savas AC, Yeh WW, Feng P. The emerging roles of glutamine amidotransferases in metabolism and immune defense. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:783-797. [PMID: 38743960 PMCID: PMC11561158 DOI: 10.1080/15257770.2024.2351135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
Glutamine amidotransferases (GATs) catalyze the synthesis of nucleotides, amino acids, glycoproteins and an enzyme cofactor, thus serving as key metabolic enzymes for cell proliferation. Carbamoyl-phosphate synthetase, Aspartate transcarbamoylase, and Dihydroorotase (CAD) is a multifunctional enzyme of the GAT family and catalyzes the first three steps of the de novo pyrimidine synthesis. Following our findings that cellular GATs are involved in immune evasion during herpesvirus infection, we discovered that CAD reprograms cellular metabolism to fuel aerobic glycolysis and nucleotide synthesis via deamidating RelA. Deamidated RelA activates the expression of key glycolytic enzymes, rather than that of the inflammatory NF-κB-responsive genes. As such, cancer cells prime RelA for deamidation via up-regulating CAD activity or accumulating RelA mutations. Interestingly, the recently emerged SARS-CoV-2 also activates CAD to couple evasion of inflammatory response to activated nucleotide synthesis. A small molecule inhibitor of CAD depletes nucleotide supply and boosts antiviral inflammatory response, thus greatly reducing SARS-CoV-2 replication. Additionally, we also found that CTP synthase 1 (CTPS1) deamidates interferon (IFN) regulatory factor 3 (IRF3) to mute IFN induction. Our previous studies have implicated phosphoribosyl formylglycinamidine synthase (PFAS) and phosphoribosyl pyrophosphate amidotransferase (PPAT) in deamidating retinoic acid-inducible gene I (RIG-I) and evading dsRNA-induced innate immune defense in herpesvirus infection. Overall, these studies have uncovered an unconventional enzymatic activity of cellular GATs in metabolism and immune defense, offering a molecular link intimately coupling these fundamental biological processes.
Collapse
Affiliation(s)
- Taolin Xie
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Chao Qin
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Ali Can Savas
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Wayne Wei Yeh
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
29
|
Li K, Fan C, Chen J, Xu X, Lu C, Shao H, Xi Y. Role of oxidative stress-induced ferroptosis in cancer therapy. J Cell Mol Med 2024; 28:e18399. [PMID: 38757920 PMCID: PMC11100387 DOI: 10.1111/jcmm.18399] [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: 10/05/2023] [Revised: 02/06/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024] Open
Abstract
Ferroptosis is a distinct mode of cell death, distinguishing itself from typical apoptosis by its reliance on the accumulation of iron ions and lipid peroxides. Cells manifest an imbalance between oxidative stress and antioxidant equilibrium during certain pathological contexts, such as tumours, resulting in oxidative stress. Notably, recent investigations propose that heightened intracellular reactive oxygen species (ROS) due to oxidative stress can heighten cellular susceptibility to ferroptosis inducers or expedite the onset of ferroptosis. Consequently, comprehending role of ROS in the initiation of ferroptosis has significance in elucidating disorders related to oxidative stress. Moreover, an exhaustive exploration into the mechanism and control of ferroptosis might offer novel targets for addressing specific tumour types. Within this context, our review delves into recent fundamental pathways and the molecular foundation of ferroptosis. Four classical ferroptotic molecular pathways are well characterized, namely, glutathione peroxidase 4-centred molecular pathway, nuclear factor erythroid 2-related factor 2 molecular pathway, mitochondrial molecular pathway, and mTOR-dependent autophagy pathway. Furthermore, we seek to elucidate the regulatory contributions enacted by ROS. Additionally, we provide an overview of targeted medications targeting four molecular pathways implicated in ferroptosis and their potential clinical applications. Here, we review the role of ROS and oxidative stress in ferroptosis, and we discuss opportunities to use ferroptosis as a new strategy for cancer therapy and point out the current challenges persisting within the domain of ROS-regulated anticancer drug research and development.
Collapse
Affiliation(s)
- Keqing Li
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science CenterNingbo UniversityNingboChina
| | - Chengjiang Fan
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science CenterNingbo UniversityNingboChina
| | - Jianing Chen
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science CenterNingbo UniversityNingboChina
| | - Xin Xu
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science CenterNingbo UniversityNingboChina
| | - Chuwei Lu
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science CenterNingbo UniversityNingboChina
| | - Hanjie Shao
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science CenterNingbo UniversityNingboChina
| | - Yang Xi
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science CenterNingbo UniversityNingboChina
| |
Collapse
|
30
|
Guo S, Miao M, Wu Y, Pan D, Wu Q, Kang Z, Zeng J, Zhong G, Liu C, Wang J. DHODH inhibition represents a therapeutic strategy and improves abiraterone treatment in castration-resistant prostate cancer. Oncogene 2024; 43:1399-1410. [PMID: 38480915 DOI: 10.1038/s41388-024-03005-4] [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: 11/19/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 05/05/2024]
Abstract
Castration-resistant prostate cancer (CRPC) is an aggressive disease with poor prognosis, and there is an urgent need for more effective therapeutic targets to address this challenge. Here, we showed that dihydroorotate dehydrogenase (DHODH), an enzyme crucial in the pyrimidine biosynthesis pathway, is a promising therapeutic target for CRPC. The transcript levels of DHODH were significantly elevated in prostate tumors and were negatively correlated with the prognosis of patients with prostate cancer. DHODH inhibition effectively suppressed CRPC progression by blocking cell cycle progression and inducing apoptosis. Notably, treatment with DHODH inhibitor BAY2402234 activated androgen biosynthesis signaling in CRPC cells. However, the combination treatment with BAY2402234 and abiraterone decreased intratumoral testosterone levels and induced apoptosis, which inhibited the growth of CWR22Rv1 xenograft tumors and patient-derived xenograft organoids. Taken together, these results establish DHODH as a key player in CRPC and as a potential therapeutic target for advanced prostate cancer.
Collapse
Affiliation(s)
- Shaoqiang Guo
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Miaomiao Miao
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yufeng Wu
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dongyue Pan
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Qinyan Wu
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhanfang Kang
- Guangdong Engineering Research Center of Urinary Continence and Reproductive Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Jianwen Zeng
- Guangdong Engineering Research Center of Urinary Continence and Reproductive Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Guoping Zhong
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Chengfei Liu
- Department of Urologic Surgery, University of California, Davis, CA, USA.
- UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA.
| | - Junjian Wang
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
31
|
Feng F, He S, Li X, He J, Luo L. Mitochondria-mediated Ferroptosis in Diseases Therapy: From Molecular Mechanisms to Implications. Aging Dis 2024; 15:714-738. [PMID: 37548939 PMCID: PMC10917537 DOI: 10.14336/ad.2023.0717] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/17/2023] [Indexed: 08/08/2023] Open
Abstract
Ferroptosis, a type of cell death involving iron and lipid peroxidation, has been found to be closely associated with the development of many diseases. Mitochondria are vital components of eukaryotic cells, serving important functions in energy production, cellular metabolism, and apoptosis regulation. Presently, the precise relationship between mitochondria and ferroptosis remains unclear. In this study, we aim to systematically elucidate the mechanisms via which mitochondria regulate ferroptosis from multiple perspectives to provide novel insights into mitochondrial functions in ferroptosis. Additionally, we present a comprehensive overview of how mitochondria contribute to ferroptosis in different conditions, including cancer, cardiovascular disease, inflammatory disease, mitochondrial DNA depletion syndrome, and novel coronavirus pneumonia. Gaining a comprehensive understanding of the involvement of mitochondria in ferroptosis could lead to more effective approaches for both basic cell biology studies and medical treatments.
Collapse
Affiliation(s)
- Fuhai Feng
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Xiaoling Li
- Animal Experiment Center, Guangdong Medical University, Zhanjiang, China.
| | - Jiake He
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, China.
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, China.
| |
Collapse
|
32
|
Liu N, Lin Q, Huang Z, Liu C, Qin J, Yu Y, Chen W, Zhang J, Jiang M, Gao X, Huo S, Zhu X. Mitochondria-Targeted Prodrug Nanoassemblies for Efficient Ferroptosis-Based Therapy via Devastating Ferroptosis Defense Systems. ACS NANO 2024; 18:7945-7958. [PMID: 38452275 DOI: 10.1021/acsnano.3c10133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Ferroptosis is a form of regulated cell death accompanied by lipid reactive oxygen species (ROS) accumulation in an iron-dependent manner. However, the efficiency of tumorous ferroptosis was seriously restricted by intracellular ferroptosis defense systems, the glutathione peroxidase 4 (GPX4) system, and the ubiquinol (CoQH2) system. Inspired by the crucial role of mitochondria in the ferroptosis process, we reported a prodrug nanoassembly capable of unleashing potent mitochondrial lipid peroxidation and ferroptotic cell death. Dihydroorotate dehydrogenase (DHODH) inhibitor (QA) was combined with triphenylphosphonium moiety through a disulfide-containing linker to engineer well-defined nanoassemblies (QSSP) within a single-molecular framework. After being trapped in cancer cells, the acidic condition provoked the structural disassembly of QSSP to liberate free prodrug molecules. The mitochondrial membrane-potential-driven accumulation of the lipophilic cation prodrug was delivered explicitly into the mitochondria. Afterward, the thiol-disulfide exchange would occur accompanied by downregulation of reduced glutathione levels, thus resulting in mitochondria-localized GPX4 inactivation for ferroptosis. Simultaneously, the released QA from the hydrolysis reaction of the adjacent ester bond could further devastate mitochondrial defense and evoke robust ferroptosis via the DHODH-CoQH2 system. This subcellular targeted nanoassembly provides a reference for designing ferroptosis-based strategy for efficient cancer therapy through interfering antiferroptosis systems.
Collapse
Affiliation(s)
- Nian Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Qian Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zhenkun Huang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Chen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Jingbo Qin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Yanlin Yu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Weibin Chen
- School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jingbo Zhang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Min Jiang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xuemin Gao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
33
|
Zhang S, Qin O, Wu S, Xu H, Huang W, Hailiang S. A pyrimidine metabolism-related signature for prognostic and immunotherapeutic response prediction in hepatocellular carcinoma by integrating analyses. Aging (Albany NY) 2024; 16:5545-5566. [PMID: 38517376 PMCID: PMC11006494 DOI: 10.18632/aging.205663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/02/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), with discouraging morbidity and mortality, ranks as one of the most prevalent tumors worldwide. Pyrimidine metabolism is a critical process that regulates DNA and RNA synthesis in cells. It is imperative to investigate the significance of pyrimidine metabolism in liver cancer. METHODS Transcriptome and clinical data were downloaded from the TCGA database and the GEO database. The genes related to pyrimidine metabolism were sourced from the MSigDB. The pyrimidine metabolism-related signature (PMRS) was constructed through Cox regression and Lasso regression and then verified in the external validation set from the ICGC database. Functional enrichment, immune infiltration analysis, drug sensitivity, and Immunophenoscore (IPS) were further implemented to predict the response to immunotherapy. The role of PMRS in the malignant phenotype of hepatocellular carcinoma was explored by conducting a series of in vitro experiments. RESULTS Our study developed a four-genes PMRS which demonstrates a substantial correlation with the prognosis of HCC patients, serving as an independent predictor in clinical practice. The result of risk-stratified analysis yielded evidence that low-risk patients experienced more favorable clinical outcomes. The nomogram exhibited remarkable prognostic predictive value. The subsequent results revealed that low-risk patients manifested a more promising response to immunotherapy. Moreover, the results of cell experiments demonstrated that the downregulation of DCK markedly inhibited the malignant phenotype of hepatocellular carcinoma. CONCLUSIONS Our pyrimidine metabolism-centered prognostic signature accurately predicts overall survival, immune status, and treatment response in hepatocellular carcinoma (HCC) patients, offering innovative insights for precise diagnosis, personalized treatment, and improved prognosis.
Collapse
Affiliation(s)
- Shihang Zhang
- Department of General Surgery, Dalang Hospital, Dongguan, Guangdong, P.R. China
| | - Ouyang Qin
- Department of General Surgery, Dalang Hospital, Dongguan, Guangdong, P.R. China
| | - Shu Wu
- Affiliated Dongguan Hospital Southern Medical University (Dongguan People’s Hospital) Dongguan Guangdong, Guangdong, P.R. China
| | - Huanming Xu
- Department of General Surgery, Dalang Hospital, Dongguan, Guangdong, P.R. China
| | - Wei Huang
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| | - Song Hailiang
- Department of General Surgery, Dalang Hospital, Dongguan, Guangdong, P.R. China
| |
Collapse
|
34
|
Zhang CH, Yan YJ, Luo Q. The molecular mechanisms and potential drug targets of ferroptosis in myocardial ischemia-reperfusion injury. Life Sci 2024; 340:122439. [PMID: 38278348 DOI: 10.1016/j.lfs.2024.122439] [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: 11/23/2023] [Revised: 01/07/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI), caused by the initial interruption and subsequent restoration of coronary artery blood, results in further damage to cardiac function, affecting the prognosis of patients with acute myocardial infarction. Ferroptosis is an iron-dependent, superoxide-driven, non-apoptotic form of regulated cell death that is involved in the pathogenesis of MIRI. Ferroptosis is characterized by the accumulation of lipid peroxides (LOOH) and redox disequilibrium. Free iron ions can induce lipid oxidative stress as a substrate of the Fenton reaction and lipoxygenase (LOX) and participate in the inactivation of a variety of lipid antioxidants including CoQ10 and GPX4, destroying the redox balance and causing cell death. The metabolism of amino acid, iron, and lipids, including associated pathways, is considered as a specific hallmark of ferroptosis. This review systematically summarizes the latest research progress on the mechanisms of ferroptosis and discusses and analyzes the therapeutic approaches targeting ferroptosis to alleviate MIRI.
Collapse
Affiliation(s)
- Chen-Hua Zhang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yu-Jie Yan
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Qi Luo
- School of Basic Medical Science, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
35
|
Liu N, Chen M. Crosstalk between ferroptosis and cuproptosis: From mechanism to potential clinical application. Biomed Pharmacother 2024; 171:116115. [PMID: 38181713 DOI: 10.1016/j.biopha.2023.116115] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/23/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
Ferroptosis and cuproptosis, regulated forms of cell death resulting from metal ion accumulation, are closely related in terms of occurrence, cell metabolism, signaling pathways, and drug resistance. Notably, it is now understood that these processes play crucial roles in regulating physiological and pathological processes, especially in tumor development. Consequently, ferroptosis and cuproptosis have gained increasing significance as potential targets for anti-cancer drug development. This article systematically outlines the molecular mechanisms and cross-talk components of both ferroptosis and cuproptosis, elucidating their impacts on cancer. Furthermore, it investigates the clinical perspective of targeted ferroptosis and cuproptosis in cancer chemotherapy, immunotherapy, and radiotherapy. Our discussion extends to a comparative analysis of nanoparticles developed based on the mechanisms of ferroptosis and cuproptosis in cancer, contrasting them with current conventional therapies. Opportunities and challenges in cancer treatment are explored, emphasizing the potential therapeutic direction of co-targeting ferroptosis and cuproptosis. The article also attempts to analyze the clinical applications of this co-targeting approach for cancer treatment while summarizing the existing barriers that require overcoming.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Minbin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
36
|
Liu P, Zhang Z, Cai Y, Li Z, Zhou Q, Chen Q. Ferroptosis: Mechanisms and role in diabetes mellitus and its complications. Ageing Res Rev 2024; 94:102201. [PMID: 38242213 DOI: 10.1016/j.arr.2024.102201] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 01/06/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Diabetes mellitus (DM) and its complications are major diseases that affect human health and pose a serious threat to global public health. Although the prevention and treatment of DM and its complications are constantly being revised, optimal treatment strategies remain unavailable. Further exploration of new anti-diabetic strategies is an arduous task. Revealing the pathological changes and molecular mechanisms of DM and its complications is the cornerstone for exploring new therapeutic strategies. Ferroptosis is a type of newly discovered iron-dependent regulated cell death. Notably, the role of ferroptosis in the occurrence, development, and pathogenesis of DM and its complications has gradually been revealed. Numerous studies have shown that ferroptosis plays an important role in the pathophysiology and pathogenesis of DM and its associated complications. The aim of this review is to discuss the known underlying mechanisms of ferroptosis, the relationship between ferroptosis and DM, and the relationship between ferroptosis as a mode of cell death and diabetic kidney disease, diabetic retinopathy, diabetic cardiomyopathy, diabetic osteoporosis, diabetes-associated cognitive dysfunction, DM-induced erectile dysfunction, and diabetic atherosclerosis.
Collapse
Affiliation(s)
- Pan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, PR China
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, Sichuan, PR China; Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan, PR China
| | - Yichen Cai
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, PR China
| | - Zhaoying Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, PR China
| | - Qian Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, PR China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, PR China.
| |
Collapse
|
37
|
Zhang J, Zhu Q, Peng Z, Li XJ, Ding PF, Gao S, Sheng B, Liu Y, Lu Y, Zhuang Z, Hang CH, Li W. Menaquinone-4 attenuates ferroptosis by upregulating DHODH through activation of SIRT1 after subarachnoid hemorrhage. Free Radic Biol Med 2024; 210:416-429. [PMID: 38042225 DOI: 10.1016/j.freeradbiomed.2023.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND Menaquinone-4(MK-4), the isoform of vitamin K2 in the brain, exerts neuroprotective effects against a variety of central nervous system disorders. This study aimed to demonstrate the anti-ferroptosis effects of MK-4 in neurons after SAH. METHODS A subarachnoid hemorrhage (SAH) model was prepared by endovascular perforation in mice. In vitro hemoglobin stimulation of primary cortical neurons mimicked SAH. MK-4, Brequinar (BQR, DHODH inhibitor), and Selisistat (SEL, SIRT1 inhibitor) were administered, respectively. Subsequently, WB, immunofluorescence was used to determine protein expression and localization, and transmission electron microscopy was used to observe neuronal mitochondrial structure while other indicators of ferroptosis were measured. RESULTS MK-4 treatment significantly upregulated the protein levels of DHODH; decreased GSH, PTGS2, NOX1, ROS, and restored mitochondrial membrane potential. Meanwhile, MK-4 upregulated the expression of SIRT1 and promoted its entry into the nucleus. BQR or SEL partially abolished the protective effect of MK-4 on, neurologic function, and ferroptosis. CONCLUSIONS Taken together, our results suggest that MK-4 attenuates ferroptosis after SAH by upregulating DHODH through the activation of SIRT1.
Collapse
Affiliation(s)
- Jiatong Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Qi Zhu
- Neurosurgical Institute, Nanjing University, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Zheng Peng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Xiao-Jian Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Peng-Fei Ding
- Neurosurgical Institute, Nanjing University, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Sen Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Bin Sheng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Yang Liu
- Neurosurgical Institute, Nanjing University, China; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
| | - Yue Lu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| |
Collapse
|
38
|
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
|
39
|
Ren X, Liu X, Hua M, Dai Y, Ren X, Sui C, Li X, Jiang Z, Tian M, Yang B. Discovery a series of novel inhibitors of human dihydroorotate dehydrogenase: Biological activity evaluation and molecular docking. Chem Biol Drug Des 2024; 103:e14388. [PMID: 37926553 DOI: 10.1111/cbdd.14388] [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: 08/27/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
Human dihydroorotate dehydrogenase (hDHODH) is a key enzyme that catalyzes the de novo synthesis of pyrimidine. In recent years, various studies have shown that inhibiting this enzyme can treat autoimmune diseases such as rheumatoid arthritis (RA) and cancer. This study designed and synthesized a series of novel thiazolidone hDHODH inhibitors. Through biological activity evaluation, Compound 14 was found to have high inhibitory activity, with an IC50 value reaching nanomolar level. Preliminary structure-activity relationship studies found that the carboxyl group in R1 and the naphthalene in R2 are key factors in improving activity. Through molecular docking, the binding mode between inhibitors and proteins was elucidated. This study provides an important reference for further optimizing hDHODH inhibitors.
Collapse
Affiliation(s)
- Xiaoli Ren
- College of Environment and Quality Inspection, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Xiaoyong Liu
- College of Environment and Quality Inspection, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Miao Hua
- Chongqing Experimental School, Chongqing, China
| | - Yan Dai
- College of Environment and Quality Inspection, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Xiaoping Ren
- College of Environment and Quality Inspection, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Chaoya Sui
- College of Environment and Quality Inspection, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Xiangbi Li
- Chongqing Auleon Biologicals Co., Ltd, Chongqing, China
| | - Zhiyong Jiang
- College of Environment and Quality Inspection, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Min Tian
- College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing, China
| | - Bing Yang
- College of Environment and Quality Inspection, Chongqing Chemical Industry Vocational College, Chongqing, China
| |
Collapse
|
40
|
Pan M, Ge CC, Niu SZ, Duan YY, Fan YM, Jin QW, Chen X, Tao JP, Huang SY. Functional analyses of Toxoplasma gondii dihydroorotase reveal a promising anti-parasitic target. FASEB J 2024; 38:e23397. [PMID: 38149908 DOI: 10.1096/fj.202301493r] [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: 07/21/2023] [Revised: 11/20/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
Toxoplasma gondii relies heavily on the de novo pyrimidine biosynthesis pathway for fueling the high uridine-5'-monophosphate (UMP) demand during parasite growth. The third step of de novo pyrimidine biosynthesis is catalyzed by dihydroorotase (DHO), a metalloenzyme that catalyzes the reversible condensation of carbamoyl aspartate to dihydroorotate. Here, functional analyses of TgDHO reveal that tachyzoites lacking DHO are impaired in overall growth due to decreased levels of UMP, and the noticeably growth restriction could be partially rescued after supplementation with uracil or high concentrations of L-dihydroorotate in vitro. When pyrimidine salvage pathway is disrupted, both DHOH35A and DHOD284E mutant strains proliferated much slower than DHO-expressing parasites, suggesting an essential role of both TgDHO His35 and Asp284 residues in parasite growth. Additionally, DHO deletion causes the limitation of bradyzoite growth under the condition of uracil supplementation or uracil deprivation. During the infection in mice, the DHO-deficient parasites are avirulent, despite the generation of smaller tissue cysts. The results reveal that TgDHO contributes to parasite growth both in vitro and in vivo. The significantly differences between TgDHO and mammalian DHO reflect that DHO can be exploited to produce specific inhibitors targeting apicomplexan parasites. Moreover, potential DHO inhibitors exert beneficial effects on enzymatic activity of TgDHO and T. gondii growth in vitro. In conclusion, these data highlight the important role of TgDHO in parasite growth and reveal that it is a promising anti-parasitic target for future control of toxoplasmosis.
Collapse
Affiliation(s)
- Ming Pan
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, PR China
| | - Ceng-Ceng Ge
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
| | - Shui-Zhu Niu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
| | - Yin-Yan Duan
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
| | - Yi-Min Fan
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
| | - Qi-Wang Jin
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, PR China
| | - Xiang Chen
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
| | - Jian-Ping Tao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
| | - Si-Yang Huang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Jiangsu Key Laboratory of Zoonosis, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, PR China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, PR China
| |
Collapse
|
41
|
Zhao S, Francois A, Kidane D. Inhibition of DHODH Enhances Replication-Associated Genomic Instability and Promotes Sensitivity in Endometrial Cancer. Cancers (Basel) 2023; 15:5727. [PMID: 38136273 PMCID: PMC10741824 DOI: 10.3390/cancers15245727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Endometrial carcinoma (EC) is the most common gynecological malignancy in the United States. De novo pyrimidine synthesis pathways generate nucleotides that are required for DNA synthesis. Approximately 38% of human endometrial tumors present with an overexpression of human dihydroorotate dehydrogenase (DHODH). However, the role of DHODH in cancer cell DNA replication and its impact on modulating a treatment response is currently unknown. Here, we report that endometrial tumors with overexpression of DHODH are associated with a high mutation count and chromosomal instability. Furthermore, tumors with an overexpression of DHODH show significant co-occurrence with mutations in DNA replication polymerases, which result in a histologically high-grade endometrial tumor. An in vitro experiment demonstrated that the inhibition of DHODH in endometrial cancer cell lines significantly induced replication-associated DNA damage and hindered replication fork progression. Furthermore, endometrial cancer cells were sensitive to the DHODH inhibitor either alone or in combination with the Poly (ADP-ribose) polymerase 1 inhibitor. Our findings may have important clinical implications for utilizing DHODH as a potential target to enhance cytotoxicity in high-grade endometrial tumors.
Collapse
Affiliation(s)
- Shengyuan Zhao
- Division of Pharmacology and Toxicology, Dell Pediatric Research Institute, College of Pharmacy, The University of Texas at Austin, 1400 Barbara Jordan Blvd. R1800, Austin, TX 78723, USA
| | - Aaliyah Francois
- Division of Pharmacology and Toxicology, Dell Pediatric Research Institute, College of Pharmacy, The University of Texas at Austin, 1400 Barbara Jordan Blvd. R1800, Austin, TX 78723, USA
| | - Dawit Kidane
- Division of Pharmacology and Toxicology, Dell Pediatric Research Institute, College of Pharmacy, The University of Texas at Austin, 1400 Barbara Jordan Blvd. R1800, Austin, TX 78723, USA
- Department of Physiology and Biophysics, College of Medicine, Howard University, 520 W Street N.W., Washington, DC 20059, USA
| |
Collapse
|
42
|
Li J, Li L, Zhang Z, Chen P, Shu H, Yang C, Chu Y, Liu J. Ferroptosis: an important player in the inflammatory response in diabetic nephropathy. Front Immunol 2023; 14:1294317. [PMID: 38111578 PMCID: PMC10725962 DOI: 10.3389/fimmu.2023.1294317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/15/2023] [Indexed: 12/20/2023] Open
Abstract
Diabetic nephropathy (DN) is a chronic inflammatory disease that affects millions of diabetic patients worldwide. The key to treating of DN is early diagnosis and prevention. Once the patient enters the clinical proteinuria stage, renal damage is difficult to reverse. Therefore, developing early treatment methods is critical. DN pathogenesis results from various factors, among which the immune response and inflammation play major roles. Ferroptosis is a newly discovered type of programmed cell death characterized by iron-dependent lipid peroxidation and excessive ROS production. Recent studies have demonstrated that inflammation activation is closely related to the occurrence and development of ferroptosis. Moreover, hyperglycemia induces iron overload, lipid peroxidation, oxidative stress, inflammation, and renal fibrosis, all of which are related to DN pathogenesis, indicating that ferroptosis plays a key role in the development of DN. Therefore, this review focuses on the regulatory mechanisms of ferroptosis, and the mutual regulatory processes involved in the occurrence and development of DN and inflammation. By discussing and analyzing the relationship between ferroptosis and inflammation in the occurrence and development of DN, we can deepen our understanding of DN pathogenesis and develop new therapeutics targeting ferroptosis or inflammation-related regulatory mechanisms for patients with DN.
Collapse
Affiliation(s)
- Jialing Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Luxin Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Zhen Zhang
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
- School of First Clinical Medical College, Mudanjiang Medical University, Mudanjiang, China
| | - Peijian Chen
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Haiying Shu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Can Yang
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Yanhui Chu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Jieting Liu
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
43
|
Wu H, Kren BT, Lane AN, Cassel TA, Higashi RM, Fan TWM, Scaria GS, Shekels LL, Klein MA, Albrecht JH. Cyclin D1 extensively reprograms metabolism to support biosynthetic pathways in hepatocytes. J Biol Chem 2023; 299:105407. [PMID: 38152849 PMCID: PMC10687208 DOI: 10.1016/j.jbc.2023.105407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 12/29/2023] Open
Abstract
Cell proliferation requires metabolic reprogramming to accommodate biosynthesis of new cell components, and similar alterations occur in cancer cells. However, the mechanisms linking the cell cycle machinery to metabolism are not well defined. Cyclin D1, along with its main partner cyclin-dependent kinase 4 (Cdk4), is a pivotal cell cycle regulator and driver oncogene that is overexpressed in many cancers. Here, we examine hepatocyte proliferation to define novel effects of cyclin D1 on biosynthetic metabolism. Metabolomic studies reveal that cyclin D1 broadly promotes biosynthetic pathways including glycolysis, the pentose phosphate pathway, and the purine and pyrimidine nucleotide synthesis in hepatocytes. Proteomic analyses demonstrate that overexpressed cyclin D1 binds to numerous metabolic enzymes including those involved in glycolysis and pyrimidine synthesis. In the glycolysis pathway, cyclin D1 activates aldolase and GAPDH, and these proteins are phosphorylated by cyclin D1/Cdk4 in vitro. De novo pyrimidine synthesis is particularly dependent on cyclin D1. Cyclin D1/Cdk4 phosphorylates the initial enzyme of this pathway, carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD), and metabolomic analysis indicates that cyclin D1 depletion markedly reduces the activity of this enzyme. Pharmacologic inhibition of Cdk4 along with the downstream pyrimidine synthesis enzyme dihydroorotate dehydrogenase synergistically inhibits proliferation and survival of hepatocellular carcinoma cells. These studies demonstrate that cyclin D1 promotes a broad network of biosynthetic pathways in hepatocytes, and this model may provide insights into potential metabolic vulnerabilities in cancer cells.
Collapse
Affiliation(s)
- Heng Wu
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Betsy T Kren
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Teresa A Cassel
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Richard M Higashi
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Teresa W M Fan
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - George S Scaria
- Hematology and Oncology Division, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Laurie L Shekels
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Mark A Klein
- Hematology and Oncology Division, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Jeffrey H Albrecht
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
44
|
Zhou X, Gou K, Xu J, Jian L, Luo Y, Li C, Guan X, Qiu J, Zou J, Zhang Y, Zhong X, Zeng T, Zhou Y, Xiao Y, Yang X, Chen W, Gao P, Liu C, Zhou Y, Tao L, Liu X, Cen X, Chen Q, Sun Q, Luo Y, Zhao Y. Discovery and Optimization of Novel hDHODH Inhibitors for the Treatment of Inflammatory Bowel Disease. J Med Chem 2023; 66:14755-14786. [PMID: 37870434 DOI: 10.1021/acs.jmedchem.3c01365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
As a key rate-limiting enzyme in the de novo synthesis of pyrimidine nucleotides, human dihydroorotate dehydrogenase (hDHODH) is considered a known target for the treatment of autoimmune diseases, including inflammatory bowel disease (IBD). Herein, BAY 41-2272 with a 1H-pyrazolo[3,4-b]pyridine scaffold was identified as an hDHODH inhibitor by screening an active compound library containing 5091 molecules. Further optimization led to 2-(1-(2-chloro-6-fluorobenzyl)-1H-pyrrolo[2,3-b]pyridin-3-yl)-5-cyclopropylpyrimidin-4-amine (w2), which was found to be the most promising and drug-like compound with potent inhibitory activity against hDHODH (IC50 = 173.4 nM). Compound w2 demonstrated acceptable pharmacokinetic characteristics and alleviated the severity of acute ulcerative colitis induced by dextran sulfate sodium in a dose-dependent manner. Notably, w2 exerted better therapeutic effects on ulcerative colitis than hDHODH inhibitor vidofludimus and Janus kinase (JAK) inhibitor tofacitinib. Taken together, w2 is a promising hDHODH inhibitor for the treatment of IBD and deserves to be developed as a preclinical candidate.
Collapse
Affiliation(s)
- Xia Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Green Pharmaceutical Technology Key Laboratory of Luzhou, Central Nervous System Drug Key Laboratory of Sichuan Province, Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Kun Gou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lunan Jian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chungen Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiao Zou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Zhang
- School of Medicine, Tibet University, Lhasa 850000, China
| | - Xi Zhong
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ting Zeng
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuzhou Xiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Weijie Chen
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ping Gao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chunqi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xingchen Liu
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingxiang Sun
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Youfu Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
45
|
Li Y, Mao K, Zang Y, Lu G, Qiu Q, Ouyang K, Zhao X, Song X, Xu L, Liang H, Qu M. Revealing the developmental characterization of rumen microbiome and its host in newly received cattle during receiving period contributes to formulating precise nutritional strategies. MICROBIOME 2023; 11:238. [PMID: 37924150 PMCID: PMC10623857 DOI: 10.1186/s40168-023-01682-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/27/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Minimizing mortality losses due to multiple stress and obtaining maximum performance are the production goals for newly received cattle. In recent years, vaccination and metaphylaxis treatment significantly decreased the mortality rate of newly received cattle, while the growth block induced by treatment is still obvious. Assessment of blood metabolites and behavior monitoring offer potential for early identification of morbid animals. Moreover, the ruminal microorganisms' homeostasis is a guarantee of beef steers' growth and health. The most critical period for newly received cattle is the first-month post-transport. Therefore, analyzing rumen metagenomics, rumen metabolomics, host metabolomics, and their interaction during receiving period (1 day before transport and at days 1/4, 16, and 30 after transport) is key to revealing the mechanism of growth retardation, and then to formulating management and nutritional practices for newly received cattle. RESULTS The levels of serum hormones (COR and ACTH), and pro-inflammatory factors (IL-1β, TNF-α, and IL-6) were highest at day 16, and lowest at day 30 after arrival. Meanwhile, the antioxidant capacity (SOD, GSH-Px, and T-AOC) was significantly decreased at day 16 and increased at day 30 after arrival. Metagenomics analysis revealed that rumen microbes, bacteria, archaea, and eukaryota had different trends among the four different time points. At day 16 post-transport, cattle had a higher abundance of ruminal bacteria and archaea than those before transport, but the eukaryote abundance was highest at day 30 post-transport. Before transport, most bacteria were mainly involved in polysaccharides digestion. At day 4 post-transport, the most significantly enriched KEGG pathways were nucleotide metabolism (pyrimidine metabolism and purine metabolism). At day 16 post-transport, the energy metabolism (glycolysis/gluconeogenesis, pyruvate metabolism) and ruminal contents of MCP and VFAs were significantly increased, but at the same time, energy loss induced by methane yields (Methanobrevibacter) together with pathogenic bacteria (Saccharopolyspora rectivirgula) were also significantly increased. At this time, the most upregulated ruminal L-ornithine produces more catabolite polyamines, which cause oxidative stress to rumen microbes and their host; the most downregulated ruminal 2',3'-cAMP provided favorable growth conditions for pathogenic bacteria, and the downregulated ruminal vitamin B6 metabolism and serum PC/LysoPC disrupt immune function and inflammation reaction. At day 30 post-transport, the ruminal L-ornithine and its catabolites (mainly spermidine and 1,3-propanediamine) were decreased, and the serum PC/LysoPC and 2',3'-cNMPs pools were increased. This is also consistent with the changes in redox, inflammation, and immune status of the host. CONCLUSIONS This study provides new ideas for regulating the health and performance of newly received cattle during the receiving period. The key point is to manage the newly received cattle about day 16 post-transport, specifically to inhibit the production of methane and polyamines, and the reproduction of harmful bacteria in the rumen, therefore improving the immunity and performance of newly received cattle. Video Abstract.
Collapse
Affiliation(s)
- Yanjiao Li
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China.
| | - Kang Mao
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Yitian Zang
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Guwei Lu
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Qinghua Qiu
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Kehui Ouyang
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Xianghui Zhao
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Xiaozhen Song
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Lanjiao Xu
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Huan Liang
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Mingren Qu
- Jiangxi Province Key Laboratory of Animal Nutrition/Animal Nutrition and Feed Safety Innovation Team, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China.
| |
Collapse
|
46
|
Ali ES, Ben-Sahra I. Regulation of nucleotide metabolism in cancers and immune disorders. Trends Cell Biol 2023; 33:950-966. [PMID: 36967301 PMCID: PMC10518033 DOI: 10.1016/j.tcb.2023.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 04/03/2023]
Abstract
Nucleotides are the foundational elements of life. Proliferative cells acquire nutrients for energy production and the synthesis of macromolecules, including proteins, lipids, and nucleic acids. Nucleotides are continuously replenished through the activation of the nucleotide synthesis pathways. Despite the importance of nucleotides in cell physiology, there is still much to learn about how the purine and pyrimidine synthesis pathways are regulated in response to intracellular and exogenous signals. Over the past decade, evidence has emerged that several signaling pathways [Akt, mechanistic target of rapamycin complex I (mTORC1), RAS, TP53, and Hippo-Yes-associated protein (YAP) signaling] alter nucleotide synthesis activity and influence cell function. Here, we examine the mechanisms by which these signaling networks affect de novo nucleotide synthesis in mammalian cells. We also discuss how these molecular links can be targeted in diseases such as cancers and immune disorders.
Collapse
Affiliation(s)
- Eunus S Ali
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
47
|
Chen Y, Guo Y, Chang H, Song Z, Wei Z, Huang Z, Zheng Z, Zhang G, Sun Y. Brequinar inhibits African swine fever virus replication in vitro by activating ferroptosis. Virol J 2023; 20:242. [PMID: 37875895 PMCID: PMC10599058 DOI: 10.1186/s12985-023-02204-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 10/04/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND African swine fever virus (ASFV) is one of the most fatal swine etiological agents and has a huge economic impact on the global pork industry. Given that no effective vaccines or anti-ASFV drugs are available, there remains a pressing need for novel anti-ASFV drugs. This study aimed to investigate the anti-African swine fever virus (ASFV) activity of brequinar, a DHODH inhibitor. METHODS The anti-ASFV activity of brequinar was investigated using IFA, HAD, HAD50, qRT-PCR, and western blotting assays. The western blotting assay was used to investigate whether brequinar inhibits ASFV replication by killing ASFV particles directly or by acting on cell factors. The confocal microscopy and western blotting assays were used to investigate whether brequinar inhibits ASFV replication by activating ferroptosis. RESULTS In this study, brequinar was found to effectively inhibit ASFV replication ex vivo in porcine alveolar macrophages (PAMs) in a dose-dependent manner. In kinetic studies, brequinar was found to maintain ASFV inhibition from 24 to 72 hpi. Mechanistically, the time-of-addition assay showed that brequinar exerted anti-ASFV activity in all treatment modes, including pre-, co-, and post-treatment rather than directly killing ASFV particles. Notably, FerroOrange, Mito-FerroGreen, and Liperfluo staining experiments showed that brequinar increased the accumulation of intracellular iron, mitochondrial iron, and lipid peroxides, respectively. Furthermore, we also found that ferroptosis agonist cisplatin treatment inhibited ASFV replication in a dose-dependent manner and the inhibitory effect of brequinar on ASFV was partially reversed by the ferroptosis inhibitor ferrostatin-1, suggesting that brequinar activates ferroptosis to inhibit ASFV replication. Interestingly, exogenous uridine supplementation attenuated the anti-ASFV activity of brequinar, indicating that brequinar inhibits ASFV replication by inhibiting DHODH activity and the depletion of intracellular pyrimidine pools; however, the induction of ferroptosis by brequinar treatment was not reversed by exogenous uridine supplementation, suggesting that brequinar activation of ferroptosis is not related to the metabolic function of pyrimidines. CONCLUSIONS Our data confirm that brequinar displays potent antiviral activity against ASFV in vitro and reveal the mechanism by which brequinar inhibits ASFV replication by activating ferroptosis, independent of inhibiting pyrimidine synthesis, providing novel targets for the development of anti-ASFV drugs.
Collapse
Affiliation(s)
- Yang Chen
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
| | - Yanchen Guo
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, Guangdong, China
| | - Hao Chang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
| | - Zebu Song
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, Guangdong, China
| | - Zhi Wei
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
| | - Zhao Huang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China
| | - Zezhong Zheng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, Guangdong, China
| | - Guihong Zhang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, Guangdong, China.
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China.
| | - Yankuo Sun
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, Guangdong, China.
| |
Collapse
|
48
|
Alexander ED, Aldridge JL, Burleson TS, Frasier CR. Teriflunomide treatment exacerbates cardiac ischemia reperfusion injury in isolated rat hearts. Cardiovasc Drugs Ther 2023; 37:1021-1026. [PMID: 35488973 PMCID: PMC9055010 DOI: 10.1007/s10557-022-07341-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 11/23/2022]
Abstract
PURPOSE Previous work suggests that Dihydroorotate dehydrogenase (DHODH) inhibition via teriflunomide (TERI) may provide protection in multiple disease models. To date, little is known about the effect of TERI on the heart. This study was performed to assess the potential effects of TERI on cardiac ischemia reperfusion injury. METHODS Male and female rat hearts were subjected to global ischemia (25 min) and reperfusion (120 min) on a Langendorff apparatus. Hearts were given either DMSO (VEH) or teriflunomide (TERI) for 5 min prior to induction of ischemia and during the reperfusion period. Left ventricular pressure, ECG, coronary flow, and infarct size were determined using established methods. Mitochondrial respiration was assessed via respirometry. RESULTS Perfusion of hearts with TERI led to no acute effects in any values measured across 500 pM-50 nM doses. However, following ischemia-reperfusion injury, we found that 50 nM TERI-treated hearts had an increase in myocardial infarction (p < 0.001). In 50 nM TERI-treated hearts, we also observed a marked increase in the severity of contracture (p < 0.001) at an earlier time-point (p = 0.004), as well as reductions in coronary flow (p = 0.037), left ventricular pressure development (p = 0.025), and the rate-pressure product (p = 0.008). No differences in mitochondrial respiration were observed with 50 nM TERI treatment (p = 0.24-0.87). CONCLUSION This study suggests that treatment with TERI leads to more negative outcomes following cardiac ischemia reperfusion, and administration of TERI to at-risk populations should receive special considerations.
Collapse
Affiliation(s)
- Emily Davis Alexander
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Jessa L Aldridge
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN, 37614, USA
| | - T Samuel Burleson
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Chad R Frasier
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
49
|
Li J, Zheng S, Fan Y, Tan K. Emerging significance and therapeutic targets of ferroptosis: a potential avenue for human kidney diseases. Cell Death Dis 2023; 14:628. [PMID: 37739961 PMCID: PMC10516929 DOI: 10.1038/s41419-023-06144-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/24/2023]
Abstract
Kidney diseases remain one of the leading causes of human death and have placed a heavy burden on the medical system. Regulated cell death contributes to the pathology of a plethora of renal diseases. Recently, with in-depth studies into kidney diseases and cell death, a new iron-dependent cell death modality, known as ferroptosis, has been identified and has attracted considerable attention among researchers in the pathogenesis of kidney diseases and therapeutics to treat them. The majority of studies suggest that ferroptosis plays an important role in the pathologies of multiple kidney diseases, such as acute kidney injury (AKI), chronic kidney disease, and renal cell carcinoma. In this review, we summarize recently identified regulatory molecular mechanisms of ferroptosis, discuss ferroptosis pathways and mechanisms of action in various kidney diseases, and describe the protective effect of ferroptosis inhibitors against kidney diseases, especially AKI. By summarizing the prominent roles of ferroptosis in different kidney diseases and the progress made in studying ferroptosis, we provide new directions and strategies for future research on kidney diseases. In summary, ferroptotic factors are potential targets for therapeutic intervention to alleviate different kidney diseases, and targeting them may lead to new treatments for patients with kidney diseases.
Collapse
Affiliation(s)
- Jinghan Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology; Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Sujuan Zheng
- Ministry of Education Key Laboratory of Molecular and Cellular Biology; Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Yumei Fan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology; Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China.
| | - Ke Tan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology; Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China.
| |
Collapse
|
50
|
Chu J, Li J, Sun L, Wei J. The Role of Cellular Defense Systems of Ferroptosis in Parkinson's Disease and Alzheimer's Disease. Int J Mol Sci 2023; 24:14108. [PMID: 37762411 PMCID: PMC10531775 DOI: 10.3390/ijms241814108] [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: 08/05/2023] [Revised: 08/31/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Parkinson's disease (PD) and Alzheimer's disease (AD) are the most common rapidly developing neurodegenerative diseases that lead to serious health and socio-economic consequences. Ferroptosis is a non-apoptotic form of cell death; there is growing evidence to support the notion that ferroptosis is involved in a variety of pathophysiological contexts, and there is increasing interest in the role of ferroptosis in PD and AD. Simultaneously, cells may have evolved four defense systems to counteract the toxic effects of ferroptosis occasioned by lipid peroxidation. This review, which focuses on the analysis of ferroptosis in the PD and AD context, outlines four cellular defense systems against ferroptosis and how each of them is involved in PD and AD.
Collapse
Affiliation(s)
- Jie Chu
- School of Physical Education, Henan University, Kaifeng 475004, China; (J.C.); (J.L.)
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Jingwen Li
- School of Physical Education, Henan University, Kaifeng 475004, China; (J.C.); (J.L.)
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Lin Sun
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, China
| | - Jianshe Wei
- School of Physical Education, Henan University, Kaifeng 475004, China; (J.C.); (J.L.)
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|