1
|
Simoneau A, Pratt CB, Wu HJ, Rajeswaran SS, Comer CG, Sudsakorn S, Zhang W, Liu S, Meier SR, Choi AH, Khendu T, Stowe H, Shen B, Whittington DA, Chen Y, Yu Y, Mallender WD, Feng T, Andersen JN, Maxwell JP, Throner S. Characterization of TNG348: A Selective, Allosteric USP1 Inhibitor That Synergizes with PARP Inhibitors in Tumors with Homologous Recombination Deficiency. Mol Cancer Ther 2025; 24:678-691. [PMID: 39886906 DOI: 10.1158/1535-7163.mct-24-0515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/28/2024] [Accepted: 01/29/2025] [Indexed: 02/01/2025]
Abstract
Inhibition of the deubiquitinating enzyme USP1 can induce synthetic lethality in tumors characterized by homologous recombination deficiency (HRD) and represents a novel therapeutic strategy for the treatment of BRCA1/2-mutant cancers, potentially including patients whose tumors have primary or acquired resistance to PARP inhibitors (PARPi). In this study, we present a comprehensive characterization of TNG348, an allosteric, selective, and reversible inhibitor of USP1. TNG348 induces dose-dependent accumulation of ubiquitinated protein substrates both in vitro and in vivo. CRISPR screens show that TNG348 exerts its antitumor effect by disrupting the translesion synthesis pathway of DNA damage tolerance through RAD18-dependent ubiquitinated PCNA. Although TNG348 and PARPi share the ability to selectively kill HRD tumor cells, CRISPR screens reveal that TNG348 and PARPi do so through discrete mechanisms. Particularly, knocking out PARP1 causes resistance to PARPi but sensitizes cells to TNG348 treatment. Consistent with these findings, combination of TNG348 with PARPi leads to synergistic antitumor effects in HRD tumors, resulting in tumor growth inhibition and regression in multiple mouse xenograft tumor models. Importantly, our data on human cancer models further show that the addition of TNG348 to PARPi treatment can overcome acquired PARPi resistance in vivo. Although the clinical development of TNG348 has been discontinued because of unexpected liver toxicity in patients (NCT06065059), the present data provide preclinical and mechanistic support for the continued exploration of USP1 as a drug target for the treatment of patients with BRCA1/2-mutant or HRD cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yi Yu
- Tango Therapeutics, Boston, Massachusetts
| | | | | | | | | | | |
Collapse
|
2
|
Throner S, Feng T, Andersen JN, Bandi M, Engel JL, Gong S, Gotur D, Gu L, Huang A, Lazarides K, Maxwell JP, McCarren P, McMillan BJ, Pham TV, Simoneau A, Tsai A, Whittington DA, Wilker E, Zhang M, Zhang W. Discovery of TNG-6132, a potent, selective, and orally bioavailable USP1 inhibitor. Bioorg Med Chem Lett 2025:130262. [PMID: 40315934 DOI: 10.1016/j.bmcl.2025.130262] [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: 03/12/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
USP1 (ubiquitin-specific peptidase 1) is a deubiquitinating enzyme that has been identified as essential in BRCA1/2 mutant cells and implicated in the DNA damage response. Inhibition of USP1 by small molecule inhibitors disrupts DNA repair and replication and is being pursued as a potential anticancer therapeutic in BRCA1/2 mutant cancers. We report the discovery of an in vitro and in vivo USP1 inhibitor tool compound TNG-6132 (18), a reversible, allosteric inhibitor of USP1, which strongly inhibits USP1 enzymatic activity. This inhibitory effect translates into in vitro cellular viability defects in a BRCA1-mutant breast cancer cell line, as well as an in vivo pharmacodynamic (PD) response and tumor growth suppression in a mouse xenograft efficacy model. Additionally, we report an X-ray co-crystal structure of TNG-6132 (18) bound in the USP1-UAF1 complex, a result that furthered our understanding of the role played by key elements of the pharmacophore of this chemotype as well as its mechanism of inhibition of USP1.
Collapse
Affiliation(s)
- Scott Throner
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Tianshu Feng
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Jannik N Andersen
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Madhavi Bandi
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Justin L Engel
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Shanzhong Gong
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Deepali Gotur
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Lina Gu
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Alan Huang
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | | | - John P Maxwell
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Patrick McCarren
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Brian J McMillan
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Truc V Pham
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Antoine Simoneau
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Alice Tsai
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | | | - Erik Wilker
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Minjie Zhang
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| | - Wenhai Zhang
- Tango Therapeutics, 201 Brookline Ave, Boston, MA 02215, United States
| |
Collapse
|
3
|
Qian C, Wang Z, Xiong Y, Zhang D, Zhong Y, Inuzuka H, Qi Y, Xie L, Chen X, Wei W, Jin J. Harnessing the Deubiquitinase USP1 for Targeted Protein Stabilization. J Am Chem Soc 2025; 147:14564-14573. [PMID: 40252079 DOI: 10.1021/jacs.5c01662] [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: 04/21/2025]
Abstract
Deubiquitinase-targeting chimera (DUBTAC) has emerged as a promising technology for targeted protein stabilization (TPS) by harnessing deubiquitinases (DUBs) to remove polyubiquitin chains from target proteins. Despite the presence of over 100 human DUBs, only OTUB1 and USP7 have been utilized in the development of DUBTAC. Hence, there is an urgent need to harness additional DUBs to expand the DUBTAC arsenal. In this work, we demonstrate for the first time that the USP1 deubiquitinase, which is overexpressed in several human cancers, can be leveraged for TPS. We report the development of novel USP1-recruiting DUBTACs by utilizing a noncovalent small-molecule inhibitor of USP1. First, we generated a USP1-based CFTR DUBTAC, MS5310, which effectively stabilized CFTR and is more potent than previously reported CFTR DUBTACs. Next, we developed first-in-class USP1-recruiting UTX DUBTACs, including MS7131, from a small-molecule inhibitor of UTX and JMJD3. Notably, MS7131 effectively stabilized the tumor suppressor UTX in a concentration- and time-dependent manner, while sparing the oncoprotein JMJD3, despite it retaining potent inhibition of JMJD3. Furthermore, UTX stabilization induced by MS7131 was dependent on the engagement of both USP1 and UTX. Consequently, MS7131, but not the parent USP1 inhibitor or UTX inhibitor, effectively reduced histone H3 lysine 27 trimethylation and significantly suppressed the proliferation and clonogenicity of cancer cells. Overall, this study highlights that USP1 can be harnessed for DUBTAC development. Moreover, we developed a valuable chemical tool, MS7131, for the investigation of UTX's distinct functions. This advancement paves the way for leveraging DUBTACs in the treatment of related diseases.
Collapse
Affiliation(s)
- Chao Qian
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Zhen Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Dingpeng Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Yue Zhong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Yihang Qi
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
4
|
Torrado C, Ashton NW, D'Andrea AD, Yap TA. USP1 inhibition: A journey from target discovery to clinical translation. Pharmacol Ther 2025; 271:108865. [PMID: 40274197 DOI: 10.1016/j.pharmthera.2025.108865] [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: 01/23/2025] [Revised: 04/04/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Ubiquitin-specific protease 1 (USP1) is a deubiquitinating enzyme involved in the DNA damage response. Upon DNA damage, USP1 stabilizes replication forks by removing monoubiquitin from PCNA and FANCD2-FANCI, thereby catalyzing critical final steps in translesion synthesis and interstrand crosslink (ICL) repair. This function is particularly crucial in BRCA1 mutant cancers, where the homologous recombination pathway is compromised, leading tumors to rely on USP1 for effective repair. USP1 is also overexpressed in BRCA1 mutant cancers, as well as other tumor types. Preclinical studies have demonstrated that knockout of USP1 is synthetically lethal in tumors with biallelic BRCA1 mutations, and this relationship is enhanced by combination with PARP inhibitors. Newly developed USP1 inhibitors have confirmed this synthetic lethality in BRCA1-deficient tumor cells. Moreover, these drugs have the potential for resensitizing platinum-resistant tumors. Currently, potent and specific USP1 inhibitors are undergoing evaluation in phase I clinical trials. RO7623066 (KSQ-4279) reported an acceptable safety profile during a phase I dose escalation study, with anemia being the most common side effect, and demonstrated robust pharmacokinetic, pharmacodynamic, and clinical activity. Other USP1 inhibitors, including SIM0501, XL309-101, and HSK39775, are currently in early clinical development. In this review, we provide an overview of the molecular function of USP1 and its importance as a therapeutic target in oncology, before focusing on the current state of preclinical and clinical development of USP1 inhibitors.
Collapse
Affiliation(s)
- Carlos Torrado
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas W Ashton
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alan D D'Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Timothy A Yap
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
5
|
Cheng B, Ding Z, Hong Y, Wang Y, Zhou Y, Chen J, Peng X, Zeng C. Research progress in DNA damage response (DDR)-targeting modulators: From hits to clinical candidates. Eur J Med Chem 2025; 287:117347. [PMID: 39908794 DOI: 10.1016/j.ejmech.2025.117347] [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: 12/25/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
In recent years, synthetic lethality has been regarded as a sound example of cancer treatment. Identifying a growing number of synthetic lethality targets has led to a substantial broadening of the application of synthetic lethality, well beyond the PAPR inhibitors employed for treating tumors with BRCA1/2 deficiencies. Especially, molecular targets within the DDR have furnished inhibitor sources and have rapidly advanced to clinical trials. In this review, we summarize the DDR-associated synthetic lethality targets such as WRN, USP1, PARP, ATR, DNA-PK, PRMT5, POLQ, and WEE1. These targets allow for the development of targeted modulators like inhibitors and degraders. Additionally, we emphasize the rational design, advantages, and potential limitations. Furthermore, we outline the promising future of DDR-targeted drug development.
Collapse
Affiliation(s)
- Binbin Cheng
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China; Department of Cardiology, Central Laboratory of Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, 323000, China
| | - Zongbao Ding
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Yimeng Hong
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China
| | - Yaping Wang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China
| | - Yingxing Zhou
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China; Huangshi Key Laboratory of Molecular Diagnosis and Individualized Treatment, Huangshi Love&health Hospital Affiliated of Hubei Polytechnic University, China.
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Xiaopeng Peng
- College of Pharmacy, Gannan Medical University, Ganzhou, 314000, China.
| | - Chunlai Zeng
- Department of Cardiology, Central Laboratory of Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, 323000, China.
| |
Collapse
|
6
|
Rennie ML, Oliver MR. Emerging frontiers in protein structure prediction following the AlphaFold revolution. J R Soc Interface 2025; 22:20240886. [PMID: 40233800 PMCID: PMC11999738 DOI: 10.1098/rsif.2024.0886] [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: 12/12/2024] [Revised: 02/04/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Models of protein structures enable molecular understanding of biological processes. Current protein structure prediction tools lie at the interface of biology, chemistry and computer science. Millions of protein structure models have been generated in a very short space of time through a revolution in protein structure prediction driven by deep learning, led by AlphaFold. This has provided a wealth of new structural information. Interpreting these predictions is critical to determining where and when this information is useful. But proteins are not static nor do they act alone, and structures of proteins interacting with other proteins and other biomolecules are critical to a complete understanding of their biological function at the molecular level. This review focuses on the application of state-of-the-art protein structure prediction to these advanced applications. We also suggest a set of guidelines for reporting AlphaFold predictions.
Collapse
|
7
|
Zhou W, Zhao Y, Qin W, Wu W, Liao C, Zhang Y, Yang X, Chen X, Wang Y, Kang Y, Wu J, Zhao J, Quan J, Wang X, Bu X, Yue X. Targeting USP1 Potentiates Radiation-Induced Type I IFN-Dependent Antitumor Immunity by Enhancing Oligo-Ubiquitinated SAR1A-Mediated STING Trafficking and Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412687. [PMID: 39976106 PMCID: PMC12005740 DOI: 10.1002/advs.202412687] [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: 10/10/2024] [Revised: 01/13/2025] [Indexed: 02/21/2025]
Abstract
The magnitude of Type I interferon (IFN) mediated innate immune response within the tumor microenvironment (TME) critically influences the effectiveness of radiotherapy. Unfortunately, due to a myriad of resistance mechanisms, the double-stranded DNA (dsDNA) signals produced by tumor cells postradiotherapy often induce a diminished response from immune cells. Through chemical screening targeting deubiquitinating enzymes, we identified USP1 (Ubiquitin Specific Peptidase 1) inhibitor as an enhancer of post-radiotherapy dsDNA responses. Mechanistically, within the context of immune-stimulatory cells in TME, USP1 serves as a suppressor in the stress-mediated stages of the cGAS (Cyclic GMP-AMP synthase) -STING (Stimulator of interferon genes protein) signaling pathway, specifically affecting the trafficking of STING from endoplasmic reticulum to Golgi apparatus. It is elucidated that SAR1A (Secretion associated Ras related GTPase 1A) requires K27-linked oligo-ubiquitination to assemble the STING-COP-II (Coat protein II) transport complex for STING trafficking. USP1 counteracts this activation by removing SAR1A ubiquitination, thereby blocking STING trafficking and activation. Consequently, pharmacological USP1 inhibition using ML323 sustains SAR1A ubiquitination and COP-II complex formation, significantly enhancing STING trafficking and subsequent Type I IFN production. This intervention substantially amplifies radiotherapy-induced immune activation in the TME, providing a strategic approach to overcome therapeutic resistance and synergize radiotherapy with immunotherapies.
Collapse
Affiliation(s)
- Weilin Zhou
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Yuxuan Zhao
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Wenjing Qin
- The First Affiliated HospitalJinan UniversityGuangzhouGuangdong510630China
| | - Weijian Wu
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Chenyang Liao
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Yiqiu Zhang
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Xingli Yang
- Department of Radiation OncologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xue Chen
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouGuangdong510257China
| | - Youqiao Wang
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Yushan Kang
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Jiaxin Wu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouGuangdong510257China
| | - Jiaojiao Zhao
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Junmin Quan
- Laboratory of Chemical OncogenomicsGuangdong Provincial Key Laboratory of Chemical GenomicsPeking University Shenzhen Graduate SchoolShenzhenGuangdong518072China
| | - Xuecen Wang
- Department of Radiation OncologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xianzhang Bu
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouGuangdong510006China
| | - Xin Yue
- The First Affiliated HospitalJinan UniversityGuangzhouGuangdong510630China
| |
Collapse
|
8
|
Chandler F, Reddy PAN, Bhutda S, Ross RL, Datta A, Walden M, Walker K, Di Donato S, Cassel JA, Prakesch MA, Aman A, Datti A, Campbell LJ, Foglizzo M, Bell L, Stein DN, Ault JR, Al-Awar RS, Calabrese AN, Sicheri F, Del Galdo F, Salvino JM, Greenberg RA, Zeqiraj E. Molecular glues that inhibit deubiquitylase activity and inflammatory signaling. Nat Struct Mol Biol 2025:10.1038/s41594-025-01517-5. [PMID: 40097626 DOI: 10.1038/s41594-025-01517-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025]
Abstract
Deubiquitylases (DUBs) are crucial in cell signaling and are often regulated by interactions within protein complexes. The BRCC36 isopeptidase complex (BRISC) regulates inflammatory signaling by cleaving K63-linked polyubiquitin chains on type I interferon receptors (IFNAR1). As a Zn2+-dependent JAMM/MPN (JAB1, MOV34, MPR1, Pad1 N-terminal) DUB, BRCC36 is challenging to target with selective inhibitors. Here, we discover first-in-class inhibitors, termed BRISC molecular glues (BLUEs), which stabilize a 16-subunit human BRISC dimer in an autoinhibited conformation, blocking active sites and interactions with the targeting subunit, serine hydroxymethyltransferase 2. This unique mode of action results in selective inhibition of BRISC over related complexes with the same catalytic subunit, splice variants and other JAMM/MPN DUBs. BLUE treatment reduced interferon-stimulated gene expression in cells containing wild-type BRISC and this effect was abolished when using structure-guided, inhibitor-resistant BRISC mutants. Additionally, BLUEs increase IFNAR1 ubiquitylation and decrease IFNAR1 surface levels, offering a potential strategy to mitigate type I interferon-mediated diseases. Our approach also provides a template for designing selective inhibitors of large protein complexes by promoting rather than blocking protein-protein interactions.
Collapse
Affiliation(s)
- Francesca Chandler
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Poli Adi Narayana Reddy
- Medicinal Chemistry, Molecular and Cellular Oncogenesis (MCO) Program and The Wistar Cancer Center Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Smita Bhutda
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca L Ross
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Arindam Datta
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Miriam Walden
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Kieran Walker
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Stefano Di Donato
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Joel A Cassel
- Medicinal Chemistry, Molecular and Cellular Oncogenesis (MCO) Program and The Wistar Cancer Center Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Michael A Prakesch
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Alessandro Datti
- Department of Agricultural, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Lisa J Campbell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Martina Foglizzo
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Lillie Bell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Daniel N Stein
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James R Ault
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Rima S Al-Awar
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Antonio N Calabrese
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Frank Sicheri
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK.
| | - Joseph M Salvino
- Medicinal Chemistry, Molecular and Cellular Oncogenesis (MCO) Program and The Wistar Cancer Center Molecular Screening, The Wistar Institute, Philadelphia, PA, USA.
| | - Roger A Greenberg
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Elton Zeqiraj
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
9
|
Zou Y, Guo T, Fu Z, Guo Z, Bo W, Yan D, Wang Q, Zeng J, Xu D, Wang T, Chen L. A structure-based framework for selective inhibitor design and optimization. Commun Biol 2025; 8:422. [PMID: 40075154 PMCID: PMC11903766 DOI: 10.1038/s42003-025-07840-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Structure-based drug design aims to create active compounds with favorable properties by analyzing target structures. Recently, deep generative models have facilitated structure-specific molecular generation. However, many methods are limited by inadequate pharmaceutical data, resulting in suboptimal molecular properties and unstable conformations. Additionally, these approaches often overlook binding pocket interactions and struggle with selective inhibitor design. To address these challenges, we developed a framework called Coarse-grained and Multi-dimensional Data-driven molecular generation (CMD-GEN). CMD-GEN bridges ligand-protein complexes with drug-like molecules by utilizing coarse-grained pharmacophore points sampled from diffusion model, enriching training data. Through a hierarchical architecture, it decomposes three-dimensional molecule generation within the pocket into pharmacophore point sampling, chemical structure generation, and conformation alignment, mitigating instability issues. CMD-GEN outperforms other methods in benchmark tests and controls drug-likeness effectively. Furthermore, CMD-GEN excels in cases across three synthetic lethal targets, and wet-lab validation with PARP1/2 inhibitors confirms its potential in selective inhibitor design.
Collapse
Affiliation(s)
- Yurong Zou
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Guo
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiyuan Fu
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongning Guo
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weichen Bo
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dengjie Yan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qiantao Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jun Zeng
- Western Health, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Carlton, VIC, Australia
| | - Dingguo Xu
- MOE Key Laboratory of Green Chemistry and Technology, College of Chemistry, Sichuan University, Chengdu, China
| | - Taijin Wang
- Chengdu Zenitar Biomedical Technology Co., Ltd., Chengdu, China.
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Chengdu Zenitar Biomedical Technology Co., Ltd., Chengdu, China.
| |
Collapse
|
10
|
Zhang J, Liu B, Ren R, Song S, Bao X, Huan X, Li H, Xu J, Yu T, Wang R, Miao ZH, Xiong B, He J, Liu T. Discovery and Optimization of a Series of Novel Morpholine-Containing USP1 Inhibitors. J Med Chem 2025; 68:3673-3699. [PMID: 39902599 DOI: 10.1021/acs.jmedchem.4c02792] [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: 02/05/2025]
Abstract
Ubiquitin-specific protease 1 (USP1), a well-known member of the deubiquitinating enzymes, serves as a key regulator in DNA damage repair (DDR) processes. Herein, we utilized ring-opening and cyclization strategies based on KSQ-4279 to design a novel series of USP1 inhibitors featuring a morpholine scaffold. Notably, compound 38-P2 exhibited a more potent enzymatic and cellular inhibition activity compared to KSQ-4279. Mechanistically, 38-P2 was characterized as a selective, reversible, and noncompetitive USP1 inhibitor. 38-P2 efficiently activated the DDR pathway, induced cell cycle arrest and cell apoptosis, and inhibited cell survival. Importantly, it enhanced the sensitivity of olaparib-resistant cells to olaparib and showed a synergetic effect with andrographolide in BRCA-proficient cancer cells. Furthermore, 38-P2 had favorable pharmacokinetic profiles and good safety properties in vitro and in vivo. In the MDA-MB-436 xenograft model, 38-P2 displayed significant, dose-dependent antitumor efficacy. Overall, these findings indicate that 38-P2 is a promising lead compound for further drug development.
Collapse
Affiliation(s)
- Junjie Zhang
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Benjin Liu
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ruyue Ren
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Shanshan Song
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xubin Bao
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiajuan Huan
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongrui Li
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Jiahao Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Ting Yu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Ruifeng Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Ze-Hong Miao
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Bing Xiong
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Jinxue He
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Tongchao Liu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, P. R. China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| |
Collapse
|
11
|
Bakkar M, Khalil S, Bhayekar K, Kushwaha ND, Samarbakhsh A, Dorandish S, Edwards H, Dou QP, Ge Y, Gavande NS. Ubiquitin-Specific Protease Inhibitors for Cancer Therapy: Recent Advances and Future Prospects. Biomolecules 2025; 15:240. [PMID: 40001543 PMCID: PMC11853158 DOI: 10.3390/biom15020240] [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: 01/20/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer management has traditionally depended on chemotherapy as the mainstay of treatment; however, recent advancements in targeted therapies and immunotherapies have offered new options. Ubiquitin-specific proteases (USPs) have emerged as promising therapeutic targets in cancer treatment due to their crucial roles in regulating protein homeostasis and various essential cellular processes. This review covers the following: (1) the structural and functional characteristics of USPs, highlighting their involvement in key cancer-related pathways, and (2) the discovery, chemical structures, mechanisms of action, and potential clinical implications of USP inhibitors in cancer therapy. Particular attention is given to the role of USP inhibitors in enhancing cancer immunotherapy, e.g., modulation of the tumor microenvironment, effect on regulatory T cell function, and influence on immune checkpoint pathways. Furthermore, this review summarizes the current progress and challenges of clinical trials involving USP inhibitors as cancer therapy. We also discuss the complexities of achieving target selectivity, the ongoing efforts to develop more specific and potent USP inhibitors, and the potential of USP inhibitors to overcome drug resistance and synergize with existing cancer treatments. We finally provide a perspective on future directions in targeting USPs, including the potential for personalized medicine based on specific gene mutations, underscoring their significant potential for enhancing cancer treatment. By elucidating their mechanisms of action, clinical progress, and potential future applications, we hope that this review could serve as a useful resource for both basic scientists and clinicians in the field of cancer therapeutics.
Collapse
Affiliation(s)
- Mohamad Bakkar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
- Division of Pediatric Hematology and Oncology, Children’s Hospital of Michigan, Detroit, MI 48201, USA
| | - Sara Khalil
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
| | - Komal Bhayekar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Narva Deshwar Kushwaha
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Amirreza Samarbakhsh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Sadaf Dorandish
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Q. Ping Dou
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Navnath S. Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
12
|
Mazloumi Aboukheili AM, Walden H. USP1 in regulation of DNA repair pathways. DNA Repair (Amst) 2025; 146:103807. [PMID: 39848025 DOI: 10.1016/j.dnarep.2025.103807] [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: 12/04/2023] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/25/2025]
Abstract
Ubiquitin-specific protease 1 (USP1) is the founding member of the family of cysteine proteases that catalyse hydrolysis of the isopeptide bond between ubiquitin and targets. USP1 is often overexpressed in various cancers, and expression levels correlate with poor prognosis. USP1 and its partner USP1-associated Factor 1 (UAF1) are required for deubiquitinating monoubiquitin signals in DNA interstrand crosslink repair, and in Translesion synthesis, among others, and both proteins are subject to multiple regulations themselves. This review covers recent findings on the mechanisms and functions of USP1 in DNA repair, its regulation, and its potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
| | - Helen Walden
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, Scotland.
| |
Collapse
|
13
|
Nespolo A, Stefenatti L, Pellarin I, Gambelli A, Rampioni Vinciguerra GL, Karimbayli J, Barozzi S, Orsenigo F, Spizzo R, Nicoloso MS, Segatto I, D’Andrea S, Bartoletti M, Lucia E, Giorda G, Canzonieri V, Puglisi F, Belletti B, Schiappacassi M, Baldassarre G, Sonego M. USP1 deubiquitinates PARP1 to regulate its trapping and PARylation activity. SCIENCE ADVANCES 2024; 10:eadp6567. [PMID: 39536107 PMCID: PMC11559621 DOI: 10.1126/sciadv.adp6567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
PARP inhibitors (PARPi) represent a game-changing treatment for patients with ovarian cancer with tumors deficient for the homologous recombination (HR) pathway treated with platinum (Pt)-based therapy. PARPi exert their cytotoxic effect by both trapping PARP1 on the damaged DNA and by restraining its enzymatic activity (PARylation). How PARP1 is recruited and trapped at the DNA damage sites and how resistance to PARPi could be overcome are still matters of investigation. Here, we described PARP1 as a substrate of the deubiquitinase USP1. At molecular level, USP1 binds PARP1 to remove its K63-linked polyubiquitination and controls PARP1 chromatin trapping and PARylation activity, regulating sensitivity to PARPi. In both Pt/PARPi-sensitive and -resistant cells, USP1/PARP1 combined blockade enhances replicative stress, DNA damage, and cell death. Our work dissected the biological interaction between USP1 and PARP1 and recommended this axis as a promising and powerful therapeutic choice for not only sensitive but also chemoresistant patients with ovarian cancer irrespective of their HR status.
Collapse
Affiliation(s)
- Anna Nespolo
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Linda Stefenatti
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Ilenia Pellarin
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Alice Gambelli
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Gian Luca Rampioni Vinciguerra
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Javad Karimbayli
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Sara Barozzi
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan (MI), Italy
| | - Fabrizio Orsenigo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan (MI), Italy
| | - Riccardo Spizzo
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Milena S. Nicoloso
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Ilenia Segatto
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Sara D’Andrea
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Michele Bartoletti
- Deparment of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Emilio Lucia
- Gynecological Surgery Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Giorgio Giorda
- Gynecological Surgery Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste (TS), Italy
| | - Fabio Puglisi
- Deparment of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
- Department of Medicine, University of Udine, Udine (UD), Italy
| | - Barbara Belletti
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Monica Schiappacassi
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Gustavo Baldassarre
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| | - Maura Sonego
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano (PN), Italy
| |
Collapse
|
14
|
Chandler F, Reddy PAN, Bhutda S, Ross RL, Datta A, Walden M, Walker K, Di Donato S, Cassel JA, Prakesch MA, Aman A, Datti A, Campbell LJ, Foglizzo M, Bell L, Stein DN, Ault JR, Al-awar RS, Calabrese AN, Sicheri F, Del Galdo F, Salvino JM, Greenberg RA, Zeqiraj E. Molecular glues that inhibit deubiquitylase activity and inflammatory signalling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611787. [PMID: 39282282 PMCID: PMC11398498 DOI: 10.1101/2024.09.07.611787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Deubiquitylases (DUBs) are crucial in cell signalling and are often regulated by interactions within protein complexes. The BRCC36 isopeptidase complex (BRISC) regulates inflammatory signalling by cleaving K63-linked polyubiquitin chains on Type I interferon receptors (IFNAR1). As a Zn2+-dependent JAMM/MPN DUB, BRCC36 is challenging to target with selective inhibitors. We discovered first-in-class inhibitors, termed BRISC molecular glues (BLUEs), which stabilise a 16-subunit BRISC dimer in an autoinhibited conformation, blocking active sites and interactions with the targeting subunit SHMT2. This unique mode of action results in selective inhibition of BRISC over related complexes with the same catalytic subunit, splice variants and other JAMM/MPN DUBs. BLUE treatment reduced interferon-stimulated gene expression in cells containing wild type BRISC, and this effect was absent when using structure-guided, inhibitor-resistant BRISC mutants. Additionally, BLUEs increase IFNAR1 ubiquitylation and decrease IFNAR1 surface levels, offering a potential new strategy to mitigate Type I interferon-mediated diseases. Our approach also provides a template for designing selective inhibitors of large protein complexes by promoting, rather than blocking, protein-protein interactions.
Collapse
Affiliation(s)
- Francesca Chandler
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Poli Adi Narayana Reddy
- The Wistar Cancer Center for Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Smita Bhutda
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca L. Ross
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Arindam Datta
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Miriam Walden
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Kieran Walker
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Stefano Di Donato
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Joel A. Cassel
- The Wistar Cancer Center for Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Michael A. Prakesch
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Alessandro Datti
- Department of Agriculture, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Lisa J. Campbell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Martina Foglizzo
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Lillie Bell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Daniel N. Stein
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James R. Ault
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Rima S. Al-awar
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Antonio N. Calabrese
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Frank Sicheri
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Joseph M. Salvino
- The Wistar Cancer Center for Molecular Screening, The Wistar Institute, Philadelphia, PA, USA
| | - Roger A. Greenberg
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elton Zeqiraj
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
15
|
Beretta GL, Costantino M, Mirra L, Pettinari P, Perego P. Deubiquitinases in Ovarian Cancer: Role in Drug Resistance and Tumor Aggressiveness. Int J Biol Sci 2024; 20:5208-5222. [PMID: 39430244 PMCID: PMC11489175 DOI: 10.7150/ijbs.100355] [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: 07/01/2024] [Accepted: 09/05/2024] [Indexed: 10/22/2024] Open
Abstract
Ovarian cancer is a lethal disease due to late diagnosis and occurrence of drug resistance that limits the efficacy of platinum-based therapy. Drug resistance mechanisms include both tumor intrinsic and tumor microenvironment-related factors. A role for deubiquitinases (DUBs) is starting to emerge in ovarian cancer. DUBs are a large family of enzymes that remove ubiquitin from target proteins and participate in processes affecting drug resistance such as DNA damage repair and apoptosis. Besides, DUBs modulate the functions of T cell populations favoring an immune suppressed microenvironment. Three DUBs are proteasome-associated, whereas the large majority are not. Among the former DUBs, USP14 has been proposed to modulate transcription factors such as Bcl6 and BACH1. In addition, RPN11/PSMD14 interferes with various processes including epithelial mesenchymal transition, also favored by non-proteasomal DUBs such as USP1 by acting on Snail. Besides, USP8 by stabilizing HER family receptors can confer drug resistance. Overall, DUBs appear to be druggable, with several inhibitors under development. Based on DUBs biological role, DUBs targeting appears promising in view of combination strategies involving different therapeutic approaches. Here, we summarize the relevance of DUBs in ovarian carcinoma and provide insights into future challenges for the treatment of this disease.
Collapse
Affiliation(s)
| | | | | | | | - Paola Perego
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| |
Collapse
|
16
|
Rennie ML, Gundogdu M, Arkinson C, Liness S, Frame S, Walden H. Structural and Biochemical Insights into the Mechanism of Action of the Clinical USP1 Inhibitor, KSQ-4279. J Med Chem 2024; 67:15557-15568. [PMID: 39190802 PMCID: PMC11403619 DOI: 10.1021/acs.jmedchem.4c01184] [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: 08/29/2024]
Abstract
DNA damage triggers cell signaling cascades that mediate repair. This signaling is frequently dysregulated in cancers. The proteins that mediate this signaling are potential targets for therapeutic intervention. Ubiquitin-specific protease 1 (USP1) is one such target, with small-molecule inhibitors already in clinical trials. Here, we use biochemical assays and cryo-electron microscopy (cryo-EM) to study the clinical USP1 inhibitor, KSQ-4279 (RO7623066), and compare this to the well-established tool compound, ML323. We find that KSQ-4279 binds to the same cryptic site of USP1 as ML323 but disrupts the protein structure in subtly different ways. Inhibitor binding drives a substantial increase in thermal stability of USP1, which may be mediated through the inhibitors filling a hydrophobic tunnel-like pocket in USP1. Our results contribute to the understanding of the mechanism of action of USP1 inhibitors at the molecular level.
Collapse
Affiliation(s)
- Martin Luke Rennie
- School of Molecular Biosciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Mehmet Gundogdu
- Ubiquigent Ltd, Dundee University Incubator, James Lindsay Place, Dundee DD1 5JJ, U.K
| | - Connor Arkinson
- School of Molecular Biosciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Steven Liness
- Ubiquigent Ltd, Dundee University Incubator, James Lindsay Place, Dundee DD1 5JJ, U.K
| | - Sheelagh Frame
- Ubiquigent Ltd, Dundee University Incubator, James Lindsay Place, Dundee DD1 5JJ, U.K
| | - Helen Walden
- School of Molecular Biosciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| |
Collapse
|
17
|
Li H, Liu BJ, Xu J, Song SS, Ba R, Zhang J, Huan XJ, Wang D, Miao ZH, Liu T, He JX, Xiong B. Design, synthesis, and biological evaluation of pyrido[2,3-d]pyrimidin-7(8H)-one derivatives as potent USP1 inhibitors. Eur J Med Chem 2024; 275:116568. [PMID: 38889606 DOI: 10.1016/j.ejmech.2024.116568] [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: 04/20/2024] [Revised: 05/24/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024]
Abstract
USP1 has emerged as a novel and potential target for drug discovery in single therapeutic agents or combination with chemotherapy and molecular targeted therapy. In this study, based on the disclosed structure of ML323 and KSQ-4279, we designed and synthesized a series of pyrido[2,3-d]pyrimidin-7(8H)-one derivatives as potent USP1 inhibitors by cyclization strategy and the systematic structure-activity relationship exploration was conducted. The representative compounds 1k, 1m and 2d displayed excellent USP1/UAF inhibition and exhibited strong antiproliferation effect in NCI-H1299 cells. Further flow cytometry analysis revealed that they could arrest breast cancer cells MDA-MB-436 in the S phase. Inhibition mechanism study of compound 1m indicated these derivatives acted as reversible and noncompetitive USP1 inhibitors. Of note, the combination of compound 1m with PARP inhibitor olaparib generated enhanced cell killing in olaparib-resistant MDA-MB-436/OP cells, and compound 1m exhibited excellent oral pharmacokinetic properties in mice. Overall, our efforts may provide a reliable basis for the development of novel USP1 inhibitor as a single therapeutic agent and in combination with PARP inhibitors.
Collapse
Affiliation(s)
- Hongrui Li
- Shenyang Pharmaceutical University, 103 Wenhua Rd, Shenyang, Liaoning, 110016, PR China; Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; Yangtze Delta Drug Advanced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China
| | - Ben-Jin Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, PR China; State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, PR China
| | - Jiahao Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; Yangtze Delta Drug Advanced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China
| | - Shan-Shan Song
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, PR China
| | - Ruixian Ba
- Shenyang Pharmaceutical University, 103 Wenhua Rd, Shenyang, Liaoning, 110016, PR China; Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; Yangtze Delta Drug Advanced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China
| | - Junjie Zhang
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, PR China
| | - Xia-Juan Huan
- University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, PR China; State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, PR China
| | - Dun Wang
- Shenyang Pharmaceutical University, 103 Wenhua Rd, Shenyang, Liaoning, 110016, PR China
| | - Ze-Hong Miao
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, PR China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, PR China; State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, PR China
| | - Tongchao Liu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China.
| | - Jin-Xue He
- University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, PR China; State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, PR China.
| | - Bing Xiong
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, PR China; Yangtze Delta Drug Advanced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China.
| |
Collapse
|
18
|
Previtali V, Bagnolini G, Ciamarone A, Ferrandi G, Rinaldi F, Myers SH, Roberti M, Cavalli A. New Horizons of Synthetic Lethality in Cancer: Current Development and Future Perspectives. J Med Chem 2024; 67:11488-11521. [PMID: 38955347 DOI: 10.1021/acs.jmedchem.4c00113] [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: 07/04/2024]
Abstract
In recent years, synthetic lethality has been recognized as a solid paradigm for anticancer therapies. The discovery of a growing number of synthetic lethal targets has led to a significant expansion in the use of synthetic lethality, far beyond poly(ADP-ribose) polymerase inhibitors used to treat BRCA1/2-defective tumors. In particular, molecular targets within DNA damage response have provided a source of inhibitors that have rapidly reached clinical trials. This Perspective focuses on the most recent progress in synthetic lethal targets and their inhibitors, within and beyond the DNA damage response, describing their design and associated therapeutic strategies. We will conclude by discussing the current challenges and new opportunities for this promising field of research, to stimulate discussion in the medicinal chemistry community, allowing the investigation of synthetic lethality to reach its full potential.
Collapse
Affiliation(s)
- Viola Previtali
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Greta Bagnolini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Andrea Ciamarone
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Giovanni Ferrandi
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Francesco Rinaldi
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Samuel Harry Myers
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Marinella Roberti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Andrea Cavalli
- Computational & Chemical Biology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
19
|
Foster BM, Wang Z, Schmidt CK. DoUBLing up: ubiquitin and ubiquitin-like proteases in genome stability. Biochem J 2024; 481:515-545. [PMID: 38572758 PMCID: PMC11088880 DOI: 10.1042/bcj20230284] [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: 12/18/2023] [Revised: 03/05/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2024]
Abstract
Maintaining stability of the genome requires dedicated DNA repair and signalling processes that are essential for the faithful duplication and propagation of chromosomes. These DNA damage response (DDR) mechanisms counteract the potentially mutagenic impact of daily genotoxic stresses from both exogenous and endogenous sources. Inherent to these DNA repair pathways is the activity of protein factors that instigate repair processes in response to DNA lesions. The regulation, coordination, and orchestration of these DDR factors is carried out, in a large part, by post-translational modifications, such as phosphorylation, ubiquitylation, and modification with ubiquitin-like proteins (UBLs). The importance of ubiquitylation and UBLylation with SUMO in DNA repair is well established, with the modified targets and downstream signalling consequences relatively well characterised. However, the role of dedicated erasers for ubiquitin and UBLs, known as deubiquitylases (DUBs) and ubiquitin-like proteases (ULPs) respectively, in genome stability is less well established, particularly for emerging UBLs such as ISG15 and UFM1. In this review, we provide an overview of the known regulatory roles and mechanisms of DUBs and ULPs involved in genome stability pathways. Expanding our understanding of the molecular agents and mechanisms underlying the removal of ubiquitin and UBL modifications will be fundamental for progressing our knowledge of the DDR and likely provide new therapeutic avenues for relevant human diseases, such as cancer.
Collapse
Affiliation(s)
- Benjamin M. Foster
- Manchester Cancer Research Centre (MCRC), Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, U.K
| | - Zijuan Wang
- Manchester Cancer Research Centre (MCRC), Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, U.K
| | - Christine K. Schmidt
- Manchester Cancer Research Centre (MCRC), Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, U.K
| |
Collapse
|
20
|
Keijzer N, Priyanka A, Stijf-Bultsma Y, Fish A, Gersch M, Sixma TK. Variety in the USP deubiquitinase catalytic mechanism. Life Sci Alliance 2024; 7:e202302533. [PMID: 38355287 PMCID: PMC10867860 DOI: 10.26508/lsa.202302533] [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: 12/14/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
The ubiquitin-specific protease (USP) family of deubiquitinases (DUBs) controls cellular ubiquitin-dependent signaling events. This generates therapeutic potential, with active-site inhibitors in preclinical and clinical studies. Understanding of the USP active site is primarily guided by USP7 data, where the catalytic triad consists of cysteine, histidine, and a third residue (third critical residue), which polarizes the histidine through a hydrogen bond. A conserved aspartate (fourth critical residue) is directly adjacent to this third critical residue. Although both critical residues accommodate catalysis in USP2, these residues have not been comprehensively investigated in other USPs. Here, we quantitatively investigate their roles in five USPs. Although USP7 relies on the third critical residue for catalysis, this residue is dispensable in USP1, USP15, USP40, and USP48, where the fourth critical residue is vital instead. Furthermore, these residues vary in importance for nucleophilic attack. The diverging catalytic mechanisms of USP1 and USP7 are independent of substrate and retained in cells for USP1. This unexpected variety of catalytic mechanisms in this well-conserved protein family may generate opportunities for selective targeting of individual USPs.
Collapse
Affiliation(s)
- Niels Keijzer
- Division of Biochemistry and Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Anu Priyanka
- Division of Biochemistry and Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Yvette Stijf-Bultsma
- Division of Biochemistry and Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Alexander Fish
- Division of Biochemistry and Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Malte Gersch
- Max Planck Institute of Molecular Physiology, Chemical Genomics Centre, Dortmund, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Titia K Sixma
- Division of Biochemistry and Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
21
|
Kim MS, Baek JH, Lee J, Sivaraman A, Lee K, Chun KH. Deubiquitinase USP1 enhances CCAAT/enhancer-binding protein beta (C/EBPβ) stability and accelerates adipogenesis and lipid accumulation. Cell Death Dis 2023; 14:776. [PMID: 38012162 PMCID: PMC10681981 DOI: 10.1038/s41419-023-06317-7] [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: 04/27/2023] [Revised: 10/31/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Dysregulation of the ubiquitin-proteasome system has been implicated in the pathogenesis of several metabolic disorders, including obesity, diabetes, and non-alcoholic fatty liver disease; however, the mechanisms controlling pathogenic metabolic disorders remain unclear. Transcription factor CCAAT/enhancer binding protein beta (C/EBPβ) regulates adipogenic genes. The study showed that the expression level of C/EBPβ is post-translationally regulated by the deubiquitinase ubiquitin-specific protease 1 (USP1) and that USP1 expression is remarkably upregulated during adipocyte differentiation and in the adipose tissue of mice fed a high-fat diet (HFD). We found that USP1 directly interacts with C/EBPβ. Knock-down of USP1 decreased C/EBPβ protein stability and increased its ubiquitination. Overexpression of USP1 regulates its protein stability and ubiquitination, whereas catalytic mutant of USP1 had no effect on them. It suggests that USP1 directly deubiquitinases C/EBPβ and increases the protein expression, leading to adipogenesis and lipid accumulation. Notably, the USP1-specific inhibitor ML323-originally developed to sensitize cancer cells to DNA-damaging agents-decreased adipocyte differentiation and lipid accumulation in 3T3-L1 cells without cytotoxicity. Oral gavage of ML323 was administered to HFD-fed mice, which showed weight loss and improvement in insulin and glucose sensitivity. Both fat mass and adipocyte size in white adipose tissues were significantly reduced by ML323 treatment, which also reduced the expression of genes involved in adipogenesis and inflammatory responses. ML323 also reduced lipid accumulation, hepatic triglycerides, free fatty acids, and macrophage infiltration in the livers of HFD-fed mice. Taken together, we suggest that USP1 plays an important role in adipogenesis by regulating C/EBPβ ubiquitination, and USP1-specific inhibitor ML323 is a potential treatment option and further study by ML323 is needed for clinical application for metabolic disorders.
Collapse
Affiliation(s)
- Myung Sup Kim
- Department of Biochemistry & Molecular Biology, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Seoul, Republic of Korea
- Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jung-Hwan Baek
- Department of Biochemistry & Molecular Biology, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Seoul, Republic of Korea
- Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - JinAh Lee
- Department of Biochemistry & Molecular Biology, Seoul, Republic of Korea
| | - Aneesh Sivaraman
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Seoul, Republic of Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Seoul, Republic of Korea.
- Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
22
|
Lemonidis K, Rennie ML, Arkinson C, Chaugule VK, Clarke M, Streetley J, Walden H. Structural and biochemical basis of interdependent FANCI-FANCD2 ubiquitination. EMBO J 2023; 42:e111898. [PMID: 36385258 PMCID: PMC9890228 DOI: 10.15252/embj.2022111898] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/03/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Di-monoubiquitination of the FANCI-FANCD2 (ID2) complex is a central and crucial step for the repair of DNA interstrand crosslinks via the Fanconi anaemia pathway. While FANCD2 ubiquitination precedes FANCI ubiquitination, FANCD2 is also deubiquitinated at a faster rate than FANCI, which can result in a FANCI-ubiquitinated ID2 complex (IUb D2). Here, we present a 4.1 Å cryo-EM structure of IUb D2 complex bound to double-stranded DNA. We show that this complex, like ID2Ub and IUb D2Ub , is also in the closed ID2 conformation and clamps on DNA. The target lysine of FANCD2 (K561) becomes fully exposed in the IUb D2-DNA structure and is thus primed for ubiquitination. Similarly, FANCI's target lysine (K523) is also primed for ubiquitination in the ID2Ub -DNA complex. The IUb D2-DNA complex exhibits deubiquitination resistance, conferred by the presence of DNA and FANCD2. ID2Ub -DNA, on the other hand, can be efficiently deubiquitinated by USP1-UAF1, unless further ubiquitination on FANCI occurs. Therefore, FANCI ubiquitination effectively maintains FANCD2 ubiquitination in two ways: it prevents excessive FANCD2 deubiquitination within an IUb D2Ub -DNA complex, and it enables re-ubiquitination of FANCD2 within a transient, closed-on-DNA, IUb D2 complex.
Collapse
Affiliation(s)
- Kimon Lemonidis
- School of Molecular Biosciences, College of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Martin L Rennie
- School of Molecular Biosciences, College of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Connor Arkinson
- School of Molecular Biosciences, College of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
- Present address:
Howard Hughes Medical InstituteUniversity of CaliforniaBerkeleyCAUSA
| | - Viduth K Chaugule
- School of Molecular Biosciences, College of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Mairi Clarke
- Scottish Centre for Macromolecular ImagingUniversity of GlasgowGlasgowUK
| | - James Streetley
- Scottish Centre for Macromolecular ImagingUniversity of GlasgowGlasgowUK
| | - Helen Walden
- School of Molecular Biosciences, College of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|