1
|
Liu S, Feng L, Wang Z. DCTPP1: A promising target in cancer therapy and prognosis through nucleotide metabolism. Drug Discov Today 2025; 30:104348. [PMID: 40180312 DOI: 10.1016/j.drudis.2025.104348] [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: 01/18/2025] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Deoxycytidine triphosphate pyrophosphatase 1 (DCTPP1) is an important deoxycytidine triphosphate (dCTP) hydrolase responsible for eliminating noncanonical dCTP and maintaining deoxyribonucleoside triphosphate (dNTP) pool homeostasis. This regulation is vital for proper DNA replication and genome stability. Emerging evidence highlights the considerable role of DCTPP1 in tumor progression, chemotherapy resistance, and prognostic prediction. Consequently, DCTPP1 has emerged as a promising nucleotide metabolism-related target for cancer therapy. In this review, we provide a comprehensive summary of the structural and cellular biological features of DCTPP1, its functions, and its role in cancer. In addition, we discuss recent advancments in small molecules targeting DCTPP1, and propose potential directions for future research.
Collapse
Affiliation(s)
- Shaoxuan Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Li Feng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Zhe Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
2
|
Feng L, Wang X, Guo X, Shi L, Su S, Li X, Wang J, Tan N, Ma Y, Wang Z. Identification of Novel Target DCTPP1 for Colorectal Cancer Therapy with the Natural Small-Molecule Inhibitors Regulating Metabolic Reprogramming. Angew Chem Int Ed Engl 2024; 63:e202402543. [PMID: 39143504 DOI: 10.1002/anie.202402543] [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: 02/04/2024] [Revised: 07/04/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors. Identification of new effective drug targets for CRC and exploration of bioactive small-molecules are clinically urgent. The human dCTP pyrophosphatase 1 (DCTPP1) is a newly identified pyrophosphatase regulating the cellular nucleotide pool but remains unexplored as potential target for CRC treatment. Here, twelve unprecedented chemical architectures terpene-nonadride heterodimers (1-12) and their monomers (13-20) were isolated from endophyte Bipolaris victoriae S27. Compounds 1-12 represented the first example of terpene-nonadride heterodimers, in which nonadride monomers of 1 and 2 were also first example of 5/6 bicyclic nonadrides. A series of assays showed that 2 could repress proliferation and induce cell cycle arrest, apoptotic and autophagic CRC cell death in vitro and in vivo. Clinical cancer samples data revealed that DCTPP1 was a novel target associated with poor survival in CRC. DCTPP1 was also identified as a new target protein of 2. Mechanically, compound 2 bound to DCTPP1, inhibited its enzymatic activity, intervened with amino acid metabolic reprogramming, and exerted anti-CRC activity. Our study demonstrates that DCTPP1 was a novel potential biomarker and therapeutic target for CRC, and 2 was the first natural anti-CRC drug candidate targeting DCTPP1.
Collapse
Affiliation(s)
- Li Feng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| | - Xinjia Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| | - Xinrui Guo
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| | - Liyuan Shi
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| | - Shihuang Su
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| | - Xinjing Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| | - Jia Wang
- School of Pharmacy, Nanjing Medical University, 211166, Nanjing, People's Republic of China
| | - Ninghua Tan
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| | - Yi Ma
- State Key Laboratory of Natural Medicines, School of Engineering, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| | - Zhe Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 211198, Nanjing, People's Republic of China
| |
Collapse
|
3
|
Mitsiou VPM, Antonaki AMN, Douka MD, Litinas KE. An Overview on the Synthesis of Lamellarins and Related Compounds with Biological Interest. Molecules 2024; 29:4032. [PMID: 39274880 PMCID: PMC11396623 DOI: 10.3390/molecules29174032] [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: 07/22/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
Lamellarins are natural products with a [3,4]-fused pyrrolocoumarin skeleton possessing interesting biological properties. More than 70 members have been isolated from diverse marine organisms, such as sponges, ascidians, mollusks, and tunicates. There is a continuous interest in the synthesis of these compounds. In this review, the synthetic strategies for the synthesis of the title compounds are presented along with their biological properties. Three routes are followed for the synthesis of lamellarins. Initially, pyrrole derivatives are the starting or intermediate compounds, and then they are fused to isoquinoline or a coumarin moiety. Second, isoquinoline is the starting compound fused to an indole moiety. In the last route, coumarins are the starting compounds, which are fused to a pyrrole moiety and an isoquinoline scaffold. The synthesis of isolamellarins, azacoumestans, isoazacoumestans, and analogues is also described. The above synthesis is achieved via metal-catalyzed cross-coupling, [3 + 2] cycloaddition, substitution, and lactonization reactions. The title compounds exhibit cytotoxic, multidrug resistance (MDR), topoisomerase I-targeted antitumor, anti-HIV, antiproliferative, anti-neurodegenerative disease, and anti-inflammatory activities.
Collapse
Affiliation(s)
- Vasiliki-Panagiota M Mitsiou
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Anastasia-Maria N Antonaki
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Matina D Douka
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos E Litinas
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
4
|
Zou M, Zhou H, Gu L, Zhang J, Fang L. Therapeutic Target Identification and Drug Discovery Driven by Chemical Proteomics. BIOLOGY 2024; 13:555. [PMID: 39194493 DOI: 10.3390/biology13080555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/07/2024] [Accepted: 07/19/2024] [Indexed: 08/29/2024]
Abstract
Throughout the human lifespan, from conception to the end of life, small molecules have an intrinsic relationship with numerous physiological processes. The investigation into small-molecule targets holds significant implications for pharmacological discovery. The determination of the action sites of small molecules provide clarity into the pharmacodynamics and toxicological mechanisms of small-molecule drugs, assisting in the elucidation of drug off-target effects and resistance mechanisms. Consequently, innovative methods to study small-molecule targets have proliferated in recent years, with chemical proteomics standing out as a vanguard development in chemical biology in the post-genomic age. Chemical proteomics can non-selectively identify unknown targets of compounds within complex biological matrices, with both probe and non-probe modalities enabling effective target identification. This review attempts to summarize methods and illustrative examples of small-molecule target identification via chemical proteomics. It delves deeply into the interactions between small molecules and human biology to provide pivotal directions and strategies for the discovery and comprehension of novel pharmaceuticals, as well as to improve the evaluation of drug safety.
Collapse
Affiliation(s)
- Mingjie Zou
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Haiyuan Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Letian Gu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Jingzi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Lei Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| |
Collapse
|
5
|
Kapidou E, Litinas KE. An Overview of the Synthesis of 3,4-Fused Pyrrolocoumarins of Biological Interest. Molecules 2024; 29:2748. [PMID: 38930816 PMCID: PMC11206682 DOI: 10.3390/molecules29122748] [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: 05/15/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
3,4-Fused pyrrolocoumarins, synthetically prepared or naturally occurring, possess interesting biological properties. In this review, the synthetic strategies for the synthesis of the title compounds are presented along with their biological activities. Two routes are followed for that synthesis. In one, the pyrrole ring is formed from coumarin derivatives, such as aminocoumarins or other coumarins. In the other approach, the pyranone moiety is built from an existing pyrrole derivative or through the simultaneous formation of coumarin and pyrrole frameworks. The above syntheses are achieved via 1,3-dipolar cycloaddition reactions, Michael reaction, aza-Claisen rearrangement reactions, multi-component reactions (MCR), as well as metal-catalyzed reactions. Pyrrolocoumarins present cytotoxic, antifungal, antibacterial, α-glucosidase inhibition, antioxidant, lipoxygenase (LOX) inhibition, and fluorescent activities, as well as benzodiazepine receptor ability.
Collapse
Affiliation(s)
| | - Konstantinos E. Litinas
- Laboratory of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
6
|
Wang Y, Chen X, Chen Q, Liu T, Wu Y, Huang L, Chen Y. Expression of human dCTP pyrophosphatase 1 (DCTPP1) and its association with cisplatin resistance characteristics in ovarian cancer. J Cell Mol Med 2024; 28:e18371. [PMID: 38686496 PMCID: PMC11058668 DOI: 10.1111/jcmm.18371] [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: 09/30/2023] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Cisplatin (DDP) resistance is a major challenge in treating ovarian cancer patients. A recently discovered enzyme called dCTP pyrophosphatase 1 (DCTPP1) has been implicated in regulating cancer characteristics, including drug responses. In this study, we aimed to understand the role of DCTPP1 in cancer progression and cisplatin response. Using publicly available databases, we analysed the expression and clinical significance of DCTPP1 in ovarian cancer. Our bioinformatics analysis confirmed that DCTPP1 is significantly overexpressed in ovarian cancer and is closely associated with tumour progression and poor prognosis after cisplatin treatment. We also found that DCTPP1 located in oxidoreductase complex and may be involved in various biological processes related to cisplatin resistance, including pyrimidine nucleotide metabolism, the P53 signalling pathway and cell cycle signalling pathways. We observed higher expression of DCTPP1 in cisplatin-resistant cells (SKOV3/DDP) and samples compared to their sensitive counterparts. Additionally, we found that DCTPP1 expression was only enhanced in SKOV3/S cells when treated with cisplatin, indicating different expression patterns of DCTPP1 in cisplatin-sensitive and cisplatin-resistant cancer cells. Our study further supports the notion that cisplatin induces intracellular reactive oxygen species (ROS) and triggers cancer cell death through excessive oxidative stress. Knocking out DCTPP1 reversed the drug resistance of ovarian cancer cells by enhancing the intracellular antioxidant stress response and accumulating ROS. Based on our research findings, we conclude that DCTPP1 has prognostic value for ovarian cancer patients, and targeting DCTPP1 may be clinically significant in overcoming cisplatin resistance in ovarian cancer.
Collapse
Affiliation(s)
- Yu Wang
- Obstetrics and Gynecology center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- School of medical laboratory and BiotechnologySouthern Medical UniversityGuangzhouChina
| | - Xiangyun Chen
- School of medical laboratory and BiotechnologySouthern Medical UniversityGuangzhouChina
| | - Qiduan Chen
- Obstetrics and Gynecology center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Tiancai Liu
- School of medical laboratory and BiotechnologySouthern Medical UniversityGuangzhouChina
| | - Yingsong Wu
- School of medical laboratory and BiotechnologySouthern Medical UniversityGuangzhouChina
| | - Liping Huang
- Obstetrics and Gynecology center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yao Chen
- School of medical laboratory and BiotechnologySouthern Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
O'Dea R, Kazi N, Hoffmann-Benito A, Zhao Z, Recknagel S, Wendrich K, Janning P, Gersch M. Molecular basis for ubiquitin/Fubi cross-reactivity in USP16 and USP36. Nat Chem Biol 2023; 19:1394-1405. [PMID: 37443395 PMCID: PMC10611586 DOI: 10.1038/s41589-023-01388-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023]
Abstract
Ubiquitin and ubiquitin-like proteins typically use distinct machineries to facilitate diverse functions. The immunosuppressive ubiquitin-like protein Fubi is synthesized as an N-terminal fusion to a ribosomal protein (Fubi-S30). Its proteolytic maturation by the nucleolar deubiquitinase USP36 is strictly required for translationally competent ribosomes. What endows USP36 with this activity, how Fubi is recognized and whether other Fubi proteases exist are unclear. Here, we report a chemical tool kit that facilitated the discovery of dual ubiquitin/Fubi cleavage activity in USP16 in addition to USP36 by chemoproteomics. Crystal structures of USP36 complexed with Fubi and ubiquitin uncover its substrate recognition mechanism and explain how other deubiquitinases are restricted from Fubi. Furthermore, we introduce Fubi C-terminal hydrolase measurements and reveal a synergistic role of USP16 in Fubi-S30 maturation. Our data highlight how ubiquitin/Fubi specificity is achieved in a subset of human deubiquitinases and open the door to a systematic investigation of the Fubi system.
Collapse
Affiliation(s)
- Rachel O'Dea
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Nafizul Kazi
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Alicia Hoffmann-Benito
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Zhou Zhao
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Sarah Recknagel
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Kim Wendrich
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Petra Janning
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Malte Gersch
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany.
| |
Collapse
|
8
|
Huang Q, Tan C, Zheng C, Meng H, Wang Z, Lin GQ, Zhang W, Chen B, He QL. DCTPP1, a reliable Q-biomarker for comprehensive evaluation of the quality of tripterygium glycoside tablets based on chemical references. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154972. [PMID: 37531903 DOI: 10.1016/j.phymed.2023.154972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/14/2023] [Accepted: 07/15/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND As first-line clinical drugs, tripterygium glycoside tablets (TGTs) often have inconsistent efficacy and toxic side effects, mainly due to inadequate quality control. Therefore, clinically relevant quality standards for TGTs are urgently required. PURPOSE Based on chemical substances and considering pharmacological efficacy, we aimed to develop an effective quality evaluation method for TGTs. METHODS Representative commercial samples of TGTs were collected from different manufacturers, and qualitative UHPLC/LTQ-Orbitrap-MS and quantitative UHPLC-MS/MS analysis methods were successfully applied to evaluate their quality similarities and differences based on their chemical properties. Then the anti-immunity, anti-inflammatory and antitumor activities of TGTs and related monomers were evaluated using Jurkat, RAW264.7, MIA PaCa-2, and PANC-1 as cellular models. Subsequently, we predicted and verified small molecule-DCTPP1 interactions via molecular docking using the established DCTPP1 enzymatic activity assay. Finally, we performed a gray relational analysis to evaluate the chemical characteristics and biological effects of TGTs produced by different manufacturers. RESULTS We collected 24 batches of TGTs (D01-D24) from 5 manufacturers (Co. A, Co. B, Co. C, Co. D, Co. E) for quality evaluation. The chemical composition analysis revealed significant differences in the substance bases of the samples. The D02, D18-D20 samples from Co. B constituted a separate group that differed from other samples, mainly in their absence of diterpenoids and triterpenoids, including triptolide, triptophenolide, and triptonide. In vitro anti-immunity, antitumor and anti-inflammatory tests using the same TGT concentration revealed that, except for D02, D18-D20, the remaining 20 samples exhibited different degrees of anti-immunity, antitumor and anti-inflammatory activity. Our experiments verified that triptolide, triptophenolide, and triptonide were all DCTPP1 inhibitors, and that TGTs generally exhibited DCTPP1 enzyme inhibitory activity. Moreover, the inhibitory activity of D02, D18-D20 samples from Co. B was much lower than that of the other samples, with a nearly tenfold difference in IC50. Further comprehensive analysis revealed a high correlation between DCTPP1 enzyme inhibition activity and the anti-immunity and antitumor and anti-inflammatory activities of these samples. CONCLUSION The established DCTPP1 enzymatic activity assay proved suitable for quantitative pharmacological and pharmaceutical analysis to complement the existing quality control system for TGTs and to evaluate their effectiveness.
Collapse
Affiliation(s)
- QinWei Huang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - ChunMei Tan
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Zhejiang Institute for Food and Drug Control, Hangzhou 310052, China
| | - Cheng Zheng
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Zhejiang Institute for Food and Drug Control, Hangzhou 310052, China
| | - Hong Meng
- NMPA Key Laboratory for Animal Alternative Testing Technology of Cosmetics, Zhejiang Institute for Food and Drug Control, Hangzhou 310052, China
| | - ZhengNan Wang
- NMPA Key Laboratory for Testing and Risk Warning of Pharmaceutical Microbiology, Zhejiang Institute for Food and Drug Control, Hangzhou 310052, China
| | - Guo-Qiang Lin
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - WenTing Zhang
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Zhejiang Institute for Food and Drug Control, Hangzhou 310052, China.
| | - BiLian Chen
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Zhejiang Institute for Food and Drug Control, Hangzhou 310052, China.
| | - Qing-Li He
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
9
|
Flegel J, Shaaban S, Jia ZJ, Schulte B, Lian Y, Krzyzanowski A, Metz M, Schneidewind T, Wesseler F, Flegel A, Reich A, Brause A, Xue G, Zhang M, Dötsch L, Stender ID, Hoffmann JE, Scheel R, Janning P, Rastinejad F, Schade D, Strohmann C, Antonchick AP, Sievers S, Moura-Alves P, Ziegler S, Waldmann H. The Highly Potent AhR Agonist Picoberin Modulates Hh-Dependent Osteoblast Differentiation. J Med Chem 2022; 65:16268-16289. [PMID: 36459434 PMCID: PMC9791665 DOI: 10.1021/acs.jmedchem.2c00956] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Indexed: 12/03/2022]
Abstract
Identification and analysis of small molecule bioactivity in target-agnostic cellular assays and monitoring changes in phenotype followed by identification of the biological target are a powerful approach for the identification of novel bioactive chemical matter in particular when the monitored phenotype is disease-related and physiologically relevant. Profiling methods that enable the unbiased analysis of compound-perturbed states can suggest mechanisms of action or even targets for bioactive small molecules and may yield novel insights into biology. Here we report the enantioselective synthesis of natural-product-inspired 8-oxotetrahydroprotoberberines and the identification of Picoberin, a low picomolar inhibitor of Hedgehog (Hh)-induced osteoblast differentiation. Global transcriptome and proteome profiling revealed the aryl hydrocarbon receptor (AhR) as the molecular target of this compound and identified a cross talk between Hh and AhR signaling during osteoblast differentiation.
Collapse
Affiliation(s)
- Jana Flegel
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Chemical Biology, Technical
University Dortmund, Dortmund 44227, Germany
| | - Saad Shaaban
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Institute of Organic Chemistry, University of Vienna Währinger Str. 38, Vienna 1090, Austria
| | - Zhi Jun Jia
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Key
Laboratory of Birth Defects and Related Diseases of Women and Children,
Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Britta Schulte
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Chemical Biology, Technical
University Dortmund, Dortmund 44227, Germany
| | - Yilong Lian
- Ludwig
Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, United
Kingdom
| | - Adrian Krzyzanowski
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Chemical Biology, Technical
University Dortmund, Dortmund 44227, Germany
| | - Malte Metz
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Tabea Schneidewind
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Chemical Biology, Technical
University Dortmund, Dortmund 44227, Germany
| | - Fabian Wesseler
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Chemical Biology, Technical
University Dortmund, Dortmund 44227, Germany
| | - Anke Flegel
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Alisa Reich
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Alexandra Brause
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Gang Xue
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Minghao Zhang
- Nuffield
Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
| | - Lara Dötsch
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Chemical Biology, Technical
University Dortmund, Dortmund 44227, Germany
| | - Isabelle D. Stender
- Protein
Chemistry Facility, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Jan-Erik Hoffmann
- Protein
Chemistry Facility, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Rebecca Scheel
- Faculty
of Chemistry, Inorganic Chemistry, Technical
University Dortmund, Dortmund 44227, Germany
| | - Petra Janning
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Fraydoon Rastinejad
- Nuffield
Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ, UK
| | - Dennis Schade
- Dept.
of Pharmaceutical & Medicinal Chemistry, Institute of Pharmacy, Christian-Albrechts-University of Kiel, Kiel 24118, Germany
| | - Carsten Strohmann
- Faculty
of Chemistry, Inorganic Chemistry, Technical
University Dortmund, Dortmund 44227, Germany
| | - Andrey P. Antonchick
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Chemical Biology, Technical
University Dortmund, Dortmund 44227, Germany
- Department
of Chemistry and Forensics, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, United Kingdom
| | - Sonja Sievers
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Compound
Management and Screening Center, Dortmund 44227, Germany
| | - Pedro Moura-Alves
- Ludwig
Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, United
Kingdom
- i3S-Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC-Instituto
de Biologia Molecular e Celular, Universidade
do Porto, 4200-135 Porto, Portugal
| | - Slava Ziegler
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
| | - Herbert Waldmann
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Dortmund 44227, Germany
- Faculty
of Chemistry, Chemical Biology, Technical
University Dortmund, Dortmund 44227, Germany
| |
Collapse
|
10
|
Davies C, Dötsch L, Ciulla MG, Hennes E, Yoshida K, Gasper R, Scheel R, Sievers S, Strohmann C, Kumar K, Ziegler S, Waldmann H. Identification of a Novel Pseudo-Natural Product Type IV IDO1 Inhibitor Chemotype. Angew Chem Int Ed Engl 2022; 61:e202209374. [PMID: 35959923 DOI: 10.1002/anie.202209374] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Indexed: 01/07/2023]
Abstract
Natural product (NP)-inspired design principles provide invaluable guidance for bioactive compound discovery. Pseudo-natural products (PNPs) are de novo combinations of NP fragments to target biologically relevant chemical space not covered by NPs. We describe the design and synthesis of apoxidoles, a novel pseudo-NP class, whereby indole- and tetrahydropyridine fragments are linked in monopodal connectivity not found in nature. Apoxidoles are efficiently accessible by an enantioselective [4+2] annulation reaction. Biological evaluation revealed that apoxidoles define a new potent type IV inhibitor chemotype of indoleamine 2,3-dioxygenase 1 (IDO1), a heme-containing enzyme considered a target for the treatment of neurodegeneration, autoimmunity and cancer. Apoxidoles target apo-IDO1, prevent heme binding and induce unique amino acid positioning as revealed by crystal structure analysis. Novel type IV apo-IDO1 inhibitors are in high demand, and apoxidoles may provide new opportunities for chemical biology and medicinal chemistry research.
Collapse
Affiliation(s)
- Caitlin Davies
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Technical University of Dortmund, Department of Chemical Biology, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Lara Dötsch
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Technical University of Dortmund, Department of Chemical Biology, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Maria Gessica Ciulla
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Current address: Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | - Elisabeth Hennes
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Technical University of Dortmund, Department of Chemical Biology, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Kei Yoshida
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Raphael Gasper
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Rebecca Scheel
- Technical University of Dortmund, Department of Inorganic Chemistry, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Sonja Sievers
- Compound Management and Screening Center (COMAS), Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Carsten Strohmann
- Technical University of Dortmund, Department of Inorganic Chemistry, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Kamal Kumar
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Current address: AiCuris Anti-infective Cures AG, Friedrich-Ebert-Str. 475, 42117, Wuppertal, Germany
| | - Slava Ziegler
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
| | - Herbert Waldmann
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany
- Technical University of Dortmund, Department of Chemical Biology, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| |
Collapse
|
11
|
Combining single-cell tracking and omics improves blood stem cell fate regulator identification. Blood 2022; 140:1482-1495. [PMID: 35820055 PMCID: PMC9523371 DOI: 10.1182/blood.2022016880] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022] Open
Abstract
Molecular programs initiating cell fate divergence (CFD) are difficult to identify. Current approaches usually compare cells long after CFD initiation, therefore missing molecular changes at its start. Ideally, single cells that differ in their CFD molecular program but are otherwise identical are compared early in CFD. This is possible in diverging sister cells, which were identical until their mother's division and thus differ mainly in CFD properties. In asymmetrically dividing cells, divergent daughter fates are prospectively committed during division, and diverging sisters can thus be identified at the start of CFD. Using asymmetrically dividing blood stem cells, we developed a pipeline (ie, trackSeq) for imaging, tracking, isolating, and transcriptome sequencing of single cells. Their identities, kinship, and histories are maintained throughout, massively improving molecular noise filtering and candidate identification. In addition to many identified blood stem CFD regulators, we offer here this pipeline for use in CFDs other than asymmetric division.
Collapse
|
12
|
Davies C, Dötsch L, Ciulla MG, Hennes E, Yoshida K, Gasper R, Scheel R, Sievers S, Strohmann C, Kumar K, Ziegler S, Waldmann H. Identification of a Novel Pseudo‐Natural Product Type IV IDO1 Inhibitor Chemotype. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Caitlin Davies
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Lara Dötsch
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Maria Gessica Ciulla
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Elisabeth Hennes
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Kei Yoshida
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Raphael Gasper
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Crystallography and Biophysics Facility GERMANY
| | - Rebecca Scheel
- Technische Universität Dortmund: Technische Universitat Dortmund Inorganic Chemistry GERMANY
| | - Sonja Sievers
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Compound Management and Screening Center GERMANY
| | - Carsten Strohmann
- Technische Universität Dortmund: Technische Universitat Dortmund Inorganic Chemistry GERMANY
| | - Kamal Kumar
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Slava Ziegler
- Max-Planck-Institut für molekulare Physiologie: Max-Planck-Institut fur molekulare Physiologie Chemical Biology GERMANY
| | - Herbert Waldmann
- Max-Planck-Institute of Molecular Physiology: Max-Planck-Institut fur molekulare Physiologie Chemical Biology Otto-Hahn-Str. 11 44227 Dortmund GERMANY
| |
Collapse
|
13
|
Jiang L, Yang Q, Gao J, Yang J, He J, Xin H, Zhang X. BK Channel Deficiency in Osteoblasts Reduces Bone Formation via the Wnt/β-Catenin Pathway. Mol Cells 2021; 44:557-568. [PMID: 34385407 PMCID: PMC8424144 DOI: 10.14348/molcells.2021.0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/06/2021] [Accepted: 06/13/2021] [Indexed: 11/27/2022] Open
Abstract
Global knockout of the BK channel has been proven to affect bone formation; however, whether it directly affects osteoblast differentiation and the mechanism are elusive. In the current study, we further investigated the role of BK channels in bone development and explored whether BK channels impacted the differentiation and proliferation of osteoblasts via the canonical Wnt signaling pathway. Our findings demonstrated that knockout of Kcnma1 disrupted the osteogenesis of osteoblasts and inhibited the stabilization of β-catenin. Western blot analysis showed that the protein levels of Axin1 and USP7 increased when Kcnma1 was deficient. Together, this study confirmed that BK ablation decreased bone mass via the Wnt/β-catenin signaling pathway. Our findings also showed that USP7 might have the ability to stabilize the activity of Axin1, which would increase the degradation of β-catenin in osteoblasts.
Collapse
Affiliation(s)
- Lan Jiang
- Department of Pharmacology, School of Pharmacy & Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Qianhong Yang
- Department of Pharmacology, School of Pharmacy & Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Jianjun Gao
- Department of Bone Metabolism, Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Jiahong Yang
- Department of Pharmacology, School of Pharmacy & Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Jiaqi He
- Department of Pharmacology, School of Pharmacy & Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy & Minhang Hospital, Fudan University, Shanghai 201203, China
- Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy & Minhang Hospital, Fudan University, Shanghai 201203, China
| |
Collapse
|
14
|
Liu Z, Wang P, Wold EA, Song Q, Zhao C, Wang C, Zhou J. Small-Molecule Inhibitors Targeting the Canonical WNT Signaling Pathway for the Treatment of Cancer. J Med Chem 2021; 64:4257-4288. [PMID: 33822624 DOI: 10.1021/acs.jmedchem.0c01799] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Canonical WNT signaling is an important developmental pathway that has attracted increased attention for anticancer drug discovery. From the production and secretion of WNT ligands, their binding to membrane receptors, and the β-catenin destruction complex to the expansive β-catenin transcriptional complex, multiple components have been investigated as drug targets to modulate WNT signaling. Significant progress in developing WNT inhibitors such as porcupine inhibitors, tankyrase inhibitors, β-catenin/coactivators, protein-protein interaction inhibitors, casein kinase modulators, DVL inhibitors, and dCTPP1 inhibitors has been made, with several candidates (e.g., LGK-974, PRI-724, and ETC-159) in human clinical trials. Herein we summarize recent progress in the drug discovery and development of small-molecule inhibitors targeting the canonical WNT pathway, focusing on their specific target proteins, in vitro and in vivo activities, physicochemical properties, and therapeutic potential. The relevant opportunities and challenges toward maintaining the balance between efficacy and toxicity in effectively targeting this pathway are also highlighted.
Collapse
Affiliation(s)
- Zhiqing Liu
- Institute of Evolution and Marine Biodiversity, College of Food Science and Technology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Eric A Wold
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Qiaoling Song
- Institute of Evolution and Marine Biodiversity, College of Food Science and Technology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Chenyang Zhao
- Institute of Evolution and Marine Biodiversity, College of Food Science and Technology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Changyun Wang
- Institute of Evolution and Marine Biodiversity, College of Food Science and Technology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| |
Collapse
|
15
|
Tong L, Zhao Q, Datan E, Lin GQ, Minn I, Pomper MG, Yu B, Romo D, He QL, Liu JO. Triptolide: reflections on two decades of research and prospects for the future. Nat Prod Rep 2021; 38:843-860. [PMID: 33146205 DOI: 10.1039/d0np00054j] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Covering: 2000 to 2020 Triptolide is a bioactive diterpene triepoxide isolated from Tripterygium wilfordii Hook F, a traditional Chinese medicinal plant whose extracts have been used as anti-inflammatory and immunosuppressive remedies for centuries. Although triptolide and its analogs exhibit potent bioactivities against various cancers, and inflammatory and autoimmune diseases, none of them has been approved to be used in the clinic. This review highlights advances in material sourcing, molecular mechanisms, clinical progress and new drug design strategies for triptolide over the past two decades, along with some prospects for the future course of development of triptolide.
Collapse
Affiliation(s)
- Lu Tong
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| | - Qunfei Zhao
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| | - Emmanuel Datan
- Department of Pharmacology, Johns Hopkins School of Medicine, 725 North Wolfe Street, Hunterian Building, Room 516, Baltimore, MD 21205, USA.
| | - Guo-Qiang Lin
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China. and CAS Key Laboratory of Synthetic Chemistry of Natural Substances, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Biao Yu
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Daniel Romo
- Department of Chemistry and Biochemistry, The CPRIT Synthesis and Drug Lead Discovery Laboratory, Baylor University, Waco, Texas 76710, USA
| | - Qing-Li He
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| | - Jun O Liu
- Department of Pharmacology, Johns Hopkins School of Medicine, 725 North Wolfe Street, Hunterian Building, Room 516, Baltimore, MD 21205, USA.
| |
Collapse
|
16
|
Wilkinson IVL, Terstappen GC, Russell AJ. Combining experimental strategies for successful target deconvolution. Drug Discov Today 2020; 25:S1359-6446(20)30373-1. [PMID: 32971235 DOI: 10.1016/j.drudis.2020.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/10/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
Investment in phenotypic drug discovery has led to increased demand for rapid and robust target deconvolution to aid successful drug development. Although methods for target identification and mechanism of action (MoA) discovery are flourishing, they typically lead to lists of putative targets. Validating which target(s) are involved in the therapeutic mechanism of a compound poses a significant challenge, requiring direct binding, target engagement, and functional studies in relevant physiological contexts. A combination of orthogonal approaches can allow target identification beyond the proteome as well as aid prioritisation for resource-intensive target validation studies.
Collapse
Affiliation(s)
- Isabel V L Wilkinson
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Georg C Terstappen
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3PQ, UK
| | - Angela J Russell
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3PQ, UK.
| |
Collapse
|
17
|
Scaletti E, Claesson M, Helleday T, Jemth AS, Stenmark P. The First Structure of an Active Mammalian dCTPase and its Complexes With Substrate Analogs and Products. J Mol Biol 2020; 432:1126-1142. [PMID: 31954130 DOI: 10.1016/j.jmb.2020.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/30/2019] [Accepted: 01/03/2020] [Indexed: 11/26/2022]
Abstract
Precise regulation of dNTPs within the cellular nucleotide pool is essential for high accuracy of DNA replication and is critical for retaining the genomic integrity. Recently, human dCTPase (deoxycytidine triphosphatase), also known as DCTPP1 (human all-alpha dCTP pyrophosphatase 1), has been revealed to be a key player in the balance of pyrimidine nucleotide concentrations within cells, with DCTPP1 deficiency causing DNA damage and genetic instability in both chromosomal and mitochondrial DNA. DCTPP1 also exhibits an additional "house cleaning" function as it has been shown to be highly active against modified cytidine triphosphates, such as 5-methyl-dCTP, which, if incorrectly incorporated into DNA can introduce undesirable epigenetic marking. To date, structural studies of mammalian dCTPase have been limited to inactive constructs, which do not provide information regarding the catalytic mechanism of this important enzyme. We present here the first structures of an active mammalian dCTPase from M. musculus in complex with the nonhydrolyzable substrate analog dCMPNPP and the products 5-Me-dCMP and dCMP. These structures provide clear insights into substrate binding and catalysis and clearly elucidate why previous structures of mammalian dCTPase were catalytically inactive. The overall structure of M. musculus dCTPase is highly similar to enzymes from the all-alpha NTP phosphohydrolase superfamily. Comparison of M. musculus dCTPase with homologs from a diverse range of mammals, including humans, shows that the residues, which contribute to substrate recognition, are entirely conserved, further supporting the importance of this enzyme in the protection of genomic integrity in mammalian cells.
Collapse
Affiliation(s)
- Emma Scaletti
- Department of Experimental Medical Science, Lund University, Lund, 221 00, Sweden; Department of Biochemistry and Biophysics, Stockholm University, Stockholm, S-106 91, Sweden
| | - Magnus Claesson
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, S-106 91, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, S-171 76, Sweden; Sheffield Cancer Centre, Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK
| | - Ann-Sofie Jemth
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, S-171 76, Sweden.
| | - Pål Stenmark
- Department of Experimental Medical Science, Lund University, Lund, 221 00, Sweden; Department of Biochemistry and Biophysics, Stockholm University, Stockholm, S-106 91, Sweden.
| |
Collapse
|
18
|
Cremosnik GS, Liu J, Waldmann H. Guided by evolution: from biology oriented synthesis to pseudo natural products. Nat Prod Rep 2020; 37:1497-1510. [DOI: 10.1039/d0np00015a] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review provides an overview and historical context to two concepts for the design of natural product-inspired compound libraries and highlights the used synthetic methodologies.
Collapse
Affiliation(s)
- Gregor S. Cremosnik
- Department of Chemical Biology
- Max-Planck-Institute of Molecular Physiology
- 44227 Dortmund
- Germany
| | - Jie Liu
- Department of Chemical Biology
- Max-Planck-Institute of Molecular Physiology
- 44227 Dortmund
- Germany
- Faculty of Chemistry and Chemical Biology
| | - Herbert Waldmann
- Department of Chemical Biology
- Max-Planck-Institute of Molecular Physiology
- 44227 Dortmund
- Germany
- Faculty of Chemistry and Chemical Biology
| |
Collapse
|