1
|
Ke ZB, Chen JY, Xue YT, Lin B, Huang Q, Huang XY, Chen DN, Chen SH, Ye XJ, Zheng QS, Wei Y, Xue XY, Xu N. Mechanical signal modulates prostate cancer immune escape by USP8-mediated ubiquitination-dependent degradation of PD-L1 and MHC-1. Cell Death Dis 2025; 16:413. [PMID: 40410130 PMCID: PMC12102395 DOI: 10.1038/s41419-025-07736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 04/30/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025]
Abstract
The tumor environment of prostate cancer (PCa) tissues of high Gleason score has been proved to be more immune suppressive and has higher extracellular matrix (ECM) stiffness, but whether ECM mechanical stiffness is the cause of higher ability of invasiveness and immune escape of PCa with high Gleason score remains uncertain. In this study, we showed that higher polyacrylamide hydrogels (PAAG) stiffness resulted in the progression and immune escape of PCa via integrin β1/FAK/YAP axis. The translocation of YAP into cell nucleus to bind to TEAD2 promoted the transcriptional activation of USP8. NBR1 could be ubiquitinated, and then degraded, via interacting with P62/SQSTM1 and through autophagy-lysosome pathway. Increased expression of USP8 promoted the abundance of NBR1 via K63-linked de-ubiquitination and PD-L1 via K48-linked de-ubiquitination in response to high PAAG stiffness. NBR1-mediated selective autophagy accelerated the degradation of MHC-1 of PCa. The USP8 inhibitor presented a potential application value in sensitizing immunotherapy of PCa. Taken together, we identified a USP8-mediated de-ubiquitination mechanism that involves in the process of high PAAG stiffness-mediated high expression of PD-L1 and low expression of MHC-1 of PCa cells, which provided a rationale of immunotherapy sensitization of PCa via USP8 inhibition.
Collapse
Affiliation(s)
- Zhi-Bin Ke
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jia-Yin Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Bin Lin
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Qi Huang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xu-Yun Huang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Dong-Ning Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Shao-Hao Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xiao-Jian Ye
- Department of Ultrasonography, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
2
|
He L, He J, Jiang T, Gong R, Wan X, Duan M, Chen Z, Cheng Y. Inhibition of UCH-L1 enhances immunotherapy efficacy in triple-negative breast cancer by stabilizing PD-L1. Eur J Pharmacol 2025; 1000:177743. [PMID: 40389130 DOI: 10.1016/j.ejphar.2025.177743] [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: 01/28/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Recent research indicates that programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors show promise in treating triple-negative breast cancer (TNBC), but their efficacy is lower than anticipated, especially when used alone. Therefore, enhancing the anti-tumor immune response strategy for TNBC is crucial. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), involved in tumor cell regulation and a potential therapeutic target, has an undefined role in TNBC immunotherapy. In this study, we explored the inverse correlation between UCH-L1 and PD-L1 in TNBC patient tissues. Through in vitro experiments, we found that UCH-L1 negatively regulates PD-L1 by stabilizing the E3 ubiquitin ligase ariadne-1 homolog (ARIH1), which promotes PD-L1 ubiquitination and degradation. Further analysis in Balb/c mice xenograft tumors showed that UCH-L1 correlates with GZMB+/CD8+ T cell infiltration in TNBC, suggesting potential synergistic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. Overall, in TNBC, UCH-L1 stabilizes ARIH1, leading to low PD-L1 expression, which may explain the limited effectiveness of immunotherapy in TNBC patients. Our mouse experiments showed improved therapeutic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. These findings offer a new avenue for immunotherapy in TNBC patients.
Collapse
Affiliation(s)
- Linhao He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Jiaying He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Rong Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Xiaoya Wan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Mingwu Duan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Zonglin Chen
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China; Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China; NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, 410008, China; Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, 410011, China.
| |
Collapse
|
3
|
Kuno S, Pakpian N, Muanprasat C. The potential role of PD-1/PD-L1 small molecule inhibitors in colorectal cancer with different mechanisms of action. Eur J Pharmacol 2025; 992:177351. [PMID: 39922421 DOI: 10.1016/j.ejphar.2025.177351] [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: 09/10/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related death worldwide, with increasing incidence in younger ages highlighting the need for new or alternative therapy, of which is immune checkpoint inhibitors. Antibody-based immune checkpoint inhibitors targeting the interaction between programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) have revolutionized cancer treatment, including CRC. However, the low response rate in CRC highlights the need for additional research and innovative therapies. Small molecule inhibitors have risen as another strategy worth exploring, considering their potential to target a wide array of PD-1/PD-L1-related pathways. This review focuses on the potential of small molecule inhibitors targeting the PD-1/PD-L1 axis in CRC. Exploring various classes of small molecule inhibitors, including those that directly block the PD-1/PD-L1 interaction and others that target upstream regulators or downstream signaling pathways involved in PD-1/PD-L1-mediated immune suppression. Additionally, modulation of post-transcriptional and post-translational processes, thereby influencing the expression, stability, or localization of PD-1/PD-L1 proteins to enhance antitumor immunity, provides a multifaceted treatment approach. By disrupting these pathways, these inhibitors can restore immune system activity against tumor cells, offering new hope for overcoming resistance and improving outcomes in CRC patients who do not respond to conventional immune checkpoint inhibitors (ICIs). Integrating these small molecules into CRC treatment strategies could represent a promising advancement in the battle against the challenging disease.
Collapse
Affiliation(s)
- Suhaibee Kuno
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Nattaporn Pakpian
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand.
| |
Collapse
|
4
|
Hu Q, Shi Y, Wang H, Bing L, Xu Z. Post-translational modifications of immune checkpoints: unlocking new potentials in cancer immunotherapy. Exp Hematol Oncol 2025; 14:37. [PMID: 40087690 PMCID: PMC11907956 DOI: 10.1186/s40164-025-00627-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/27/2025] [Indexed: 03/17/2025] Open
Abstract
Immunotherapy targeting immune checkpoints has gained traction across various cancer types in clinical settings due to its notable advantages. Despite this, the overall response rates among patients remain modest, alongside issues of drug resistance and adverse effects. Hence, there is a pressing need to enhance immune checkpoint blockade (ICB) therapies. Post-translational modifications (PTMs) are crucial for protein functionality. Recent research emphasizes their pivotal role in immune checkpoint regulation, directly impacting the expression and function of these key proteins. This review delves into the influence of significant PTMs-ubiquitination, phosphorylation, and glycosylation-on immune checkpoint signaling. By targeting these modifications, novel immunotherapeutic strategies have emerged, paving the way for advancements in optimizing immune checkpoint blockade therapies in the future.
Collapse
Affiliation(s)
- Qiongjie Hu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang Province, China
- The Third Affiliated Hospital of Zhejiang, Chinese Meical University, Hangzhou, 310013, China
| | - Yueli Shi
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang Province, China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Huang Wang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liuwen Bing
- The Third Affiliated Hospital of Zhejiang, Chinese Meical University, Hangzhou, 310013, China.
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang Province, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China.
| |
Collapse
|
5
|
Huang Y, Gao Y, Lin Z, Miao H. Involvement of the ubiquitin-proteasome system in the regulation of the tumor microenvironment and progression. Genes Dis 2025; 12:101240. [PMID: 39759114 PMCID: PMC11697063 DOI: 10.1016/j.gendis.2024.101240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/11/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2025] Open
Abstract
The tumor microenvironment is a complex environment comprising tumor cells, non-tumor cells, and other critical non-cellular components. Some studies about tumor microenvironment have recently achieved remarkable progress in tumor treatment. As a substantial part of post-translational protein modification, ubiquitination is a crucial player in maintaining protein stability in cell signaling, cell growth, and a series of cellular life activities, which are also essential for regulating tumor cells or other non-tumor cells in the tumor microenvironment. This review focuses on the role and function of ubiquitination and deubiquitination modification in the tumor microenvironment while discussing the prospect of developing inhibitors targeting ubiquity-related enzymes, thereby providing ideas for future research in cancer therapy.
Collapse
Affiliation(s)
- Yulan Huang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yuan Gao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
6
|
Ge X, Zhang K, Zhu J, Chen Y, Wang Z, Wang P, Xu P, Yao J. Targeting protein modification: a new direction for immunotherapy of pancreatic cancer. Int J Biol Sci 2025; 21:63-74. [PMID: 39744438 PMCID: PMC11667816 DOI: 10.7150/ijbs.101861] [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: 08/03/2024] [Accepted: 11/03/2024] [Indexed: 01/11/2025] Open
Abstract
Post-translational modifications (PTMs) alter protein conformation by covalently attaching functional groups to substrates, influencing their biological activity, mechanisms of action, and functional performance. PTMs and their interactions are essential to many critical signal transduction processes, including tumor transformation, cancer progression, and metastasis in pancreatic cancer. Additionally, advancements in tumor immunotherapy indicate that PTMs are essential in immune cell activation, transport, and energy metabolism. This study aimed to investigate the effects of different PTMs on immunotherapy for pancreatic cancer, providing new perspectives and suggesting directions for future research.
Collapse
Affiliation(s)
- Xinyu Ge
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Ke Zhang
- The Yangzhou School of Clinical Medicine of Dalian Medical University, Jiangsu 225000, China
| | - Jie Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Yuan Chen
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Zhengwang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Peng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Peng Xu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| | - Jie Yao
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu 225000, China
| |
Collapse
|
7
|
Xian Y, Ye J, Tang Y, Zhang N, Peng C, Huang W, He G. Deubiquitinases as novel therapeutic targets for diseases. MedComm (Beijing) 2024; 5:e70036. [PMID: 39678489 PMCID: PMC11645450 DOI: 10.1002/mco2.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024] Open
Abstract
Deubiquitinating enzymes (DUBs) regulate substrate ubiquitination by removing ubiquitin or cleaving within ubiquitin chains, thereby maintaining cellular homeostasis. Approximately 100 DUBs in humans counteract E3 ubiquitin ligases, finely balancing ubiquitination and deubiquitination processes to maintain cellular proteostasis and respond to various stimuli and stresses. Given their role in modulating ubiquitination levels of various substrates, DUBs are increasingly linked to human health and disease. Here, we review the DUB family, highlighting their distinctive structural characteristics and chain-type specificities. We show that DUB family members regulate key signaling pathways, such as NF-κB, PI3K/Akt/mTOR, and MAPK, and play crucial roles in tumorigenesis and other diseases (neurodegenerative disorders, cardiovascular diseases, inflammatory disorders, and developmental diseases), making them promising therapeutic targets Our review also discusses the challenges in developing DUB inhibitors and underscores the critical role of the DUBs in cellular signaling and cancer. This comprehensive analysis enhances our understanding of the complex biological functions of the DUBs and underscores their therapeutic potential.
Collapse
Affiliation(s)
- Yali Xian
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Jing Ye
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Yu Tang
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Gu He
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
8
|
Gao Q, Li N, Pan Y, Chu P, Zhou Y, Jia H, Cheng Y, Xue G, Song J, Zhang Y, Zhu H, Sun J, Zhang B, Sun Z, Fang D. Hepatocyte growth factor promotes melanoma metastasis through ubiquitin-specific peptidase 22-mediated integrins upregulation. Cancer Lett 2024; 604:217196. [PMID: 39222676 PMCID: PMC11542356 DOI: 10.1016/j.canlet.2024.217196] [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: 01/22/2024] [Revised: 07/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Hepatocyte growth factor (HGF) plays a critical role in promoting tumor migration, invasion, and metastasis, partly by upregulating integrins. The molecular mechanisms behind how HGF facilitates integrin-mediated tumorigenesis are not fully understood. In this study, we demonstrate that the ubiquitin-specific peptidase 22 (USP22) is essential for HGF-induced melanoma metastasis. HGF treatment dramatically increased the expression of both USP22 and multiple integrin family members in particular ITGAV, ITGB3, and ITGA1. An unbiased analysis of the TCGA database reveals integrins as common downstream targets of both USP22 and HGF across multiple human cancer types. Notably, CRISPR-mediated deletion of USP22 completely eliminates HGF-induced integrin expression in melanoma cells. At the molecular level, USP22 acts as a bona fide deubiquitinase for Sp1, a transcription factor for the ITGAV, ITGB3, and ITGA1 genes. USP22 interacts with and inhibits Sp1 ubiquitination, protecting against Sp1 proteasomal degradation. Supporting this, immunohistology analysis detects a positive correlation among USP22, Sp1, and integrin αv in human melanoma tissues. This study identifies the death from the signature gene USP22 as a critical positive regulator for HGF-induced integrin expression by deubiquitinating the Sp1 transcription factor during melanoma metastasis.
Collapse
Affiliation(s)
- Qiong Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Na Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yujie Pan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Peng Chu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yuanzhang Zhou
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Huijun Jia
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yang Cheng
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guoqing Xue
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Jiankun Song
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yue Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Houyu Zhu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Jia Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Bin Zhang
- Department of Medicine, Hematology/Oncology Division, Robert H. Lurie Comprehensive Cancer Center, USA
| | - Zhaolin Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China.
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Medicine, Hematology/Oncology Division, Robert H. Lurie Comprehensive Cancer Center, USA.
| |
Collapse
|
9
|
Zhang Y, Huang Y, Yu D, Xu M, Hu H, Zhang Q, Cai M, Geng X, Zhang H, Xia J, Guo M, Lu D, Xu H, Li L, Zhang X, Wang Q, Liu S, Zhang W. Demethylzeylasteral induces PD-L1 ubiquitin-proteasome degradation and promotes antitumor immunity via targeting USP22. Acta Pharm Sin B 2024; 14:4312-4328. [PMID: 39525573 PMCID: PMC11544192 DOI: 10.1016/j.apsb.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/11/2024] [Accepted: 07/25/2024] [Indexed: 11/16/2024] Open
Abstract
Programmed cell death ligand-1 (PD-L1) is a T cell inhibitory immune checkpoint molecule that interacts with programmed cell death-1 (PD-1) to promote immune escape of tumor cells. Compared with antibody therapies, small molecule drugs show better prospects due to their advantages such as higher bioavailability, better tissue penetration, and reduced risk of immunogenicity. Here, we found that the small molecule demethylzeylasteral (Dem) can significantly downregulate the expression of PD-L1 in colorectal cancer cells and enhance the killing effect of T cells on tumor cells. Mechanistically, Dem binds to the deubiquitinating enzyme USP22 and promotes its degradation, resulting in increased ubiquitination and degradation of PD-L1 through the proteasome pathway. In addition, Dem increased the activity of cytotoxic T cells and reduced the number of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) in tumor-infiltrating lymphocytes (TILs), thereby activating the tumor immune microenvironment and inhibiting the growth of subcutaneous MC38 tumors in C57BL/6 mice. Moreover, we also found that the combination of Dem and CTLA4 antibodies can further improve the efficacy of antitumor therapy. Our study reveals the mechanism by which Dem promotes PD-L1 degradation and suggests that the combination of Dem and CTLA4 antibodies may improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Yanyan Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yun Huang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dianping Yu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mengting Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongmei Hu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qing Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Minchen Cai
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiangxin Geng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongwei Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianhua Xia
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mengmeng Guo
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dong Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hanchi Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Linyang Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xing Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qun Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
10
|
Wu Y, Mohd Sani SB, Peng K, Lin T, Tan C, Huang X, Li Z. Research progress of the Otubains subfamily in hepatocellular carcinoma. Biomed Pharmacother 2024; 179:117348. [PMID: 39208669 DOI: 10.1016/j.biopha.2024.117348] [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: 05/14/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
In cancer research, oncogenesis can be affected by modulating the deubiquitination pathway. Ubiquitination regulates proteins post-translationally in variety of physiological processes. The Otubain Subfamily includes OTUB1 (ovarian tumor-associated proteinase B1) and OTUB2(ovarian tumor-associated proteinase B2). They are deubiquitinating enzymes, which are research hotspots in tumor immunotherapy, with their implications extending across the spectrum of tumor development. Understanding their important role in tumorigenesis, includ-ing hepatocellular carcinoma (HCC) is crucial. HCC has alarming global incidence rates and mortality statistics, ranking among the top five prevalent cancers in Malaysia1. Numerous studies have consistently indicated significant expression of OTUB1 and OTUB2 in HCC cells. In addition, OTUB1 has important biological functions in cancer, suggesting its important role in tumorigenesis. However, the mechanism underlying the action of OTUB1 and OTUB2 in liver cancer remains inadequately explored. Therefore, Otubain Subfamily, as potential molecular target, holds promise for advancing HCC treatments. However, further clinical studies are required to verify its efficacy and application prospects.
Collapse
Affiliation(s)
- Yanming Wu
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Kepala Batas, Penang 13200, Malaysia.
| | - Sa'udah Badriah Mohd Sani
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Kepala Batas, Penang 13200, Malaysia.
| | - Ke Peng
- Department of Neurology, School of Clinical Medicine, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, China.
| | - Tao Lin
- Department of General Surgery, Anyang People's Hospital, Anyang, Henan 450000, China.
| | - Chenghao Tan
- Department of Social Science, Universiti Sain Malaysia, Gelugor, Penang 11700, Malaysia.
| | | | - Zhengrui Li
- Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China.
| |
Collapse
|
11
|
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
|
12
|
Wang Y, Li S, Wang W. The ubiquitin-proteasome system in the tumor immune microenvironment: a key force in combination therapy. Front Immunol 2024; 15:1436174. [PMID: 39315102 PMCID: PMC11416925 DOI: 10.3389/fimmu.2024.1436174] [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: 05/21/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a crucial role in modulating the proliferation, activation, and normal functioning of immune cells through the regulation of protein degradation and function. By influencing the expression of immune checkpoint-associated proteins, the UPS modulates T cell-mediated anti-tumor immune responses and can potentially facilitate the immune escape of tumor cells. Additionally, the UPS contributes to the remodeling of the tumor immunosuppressive microenvironment (TIME) by regulating B cells, dendritic cells (DCs), macrophages, and Treg cells. Targeting the UPS in conjunction with immune checkpoint-associated proteins, and combining these with other therapeutic approaches, may significantly enhance the efficacy of combination therapies and pave the way for novel cancer treatment strategies. In this review, we first summarize the composition and alterations of the TIME, with a particular emphasis on the role of the UPS in TIME and its interactions with various immune cell types. Finally, we explore the potential of combining UPS-targeted therapies with immunotherapy to substantially improve the effectiveness of immunotherapy and enhance patient survival outcomes.
Collapse
Affiliation(s)
- Yongmei Wang
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Saisai Li
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenqin Wang
- School of Life Sciences, Shandong University, Qingdao, Shandong, China
| |
Collapse
|
13
|
Li Z, Yu X, Yuan Z, Li L, Yin P. New horizons in the mechanisms and therapeutic strategies for PD-L1 protein degradation in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189152. [PMID: 38992509 DOI: 10.1016/j.bbcan.2024.189152] [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: 04/03/2024] [Revised: 06/12/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Programmed death-ligand 1 (PD-L1) has become a crucial focus in cancer immunotherapy considering it is found in many different cells. Cancer cells enhance the suppressive impact of programmed death receptor 1 (PD-1) through elevating PD-L1 expression, which allows them to escape immune detection. Although there have been significant improvements, the effectiveness of anti-PD-1/PD-L1 treatment is still limited to a specific group of patients. An important advancement in cancer immunotherapy involves improving the PD-L1 protein degradation. This review thoroughly examined the processes by which PD-L1 breaks down, including the intracellular pathways of ubiquitination-proteasome and autophagy-lysosome. In addition, the analysis revealed changes that affect PD-L1 stability, such as phosphorylation and glycosylation. The significant consequences of these procedures on cancer immunotherapy and their potential role in innovative therapeutic approaches are emphasised. Our future efforts will focus on understanding new ways in which PD-L1 degradation is controlled and developing innovative treatments, such as proteolysis-targeting chimeras designed specifically to degrade PD-L1. It is crucial to have a thorough comprehension of these pathways in order to improve cancer immunotherapy strategies and hopefully improve therapeutic effectiveness.
Collapse
Affiliation(s)
- Zhi Li
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Department of General surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xi Yu
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Zeting Yuan
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| | - Lei Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| |
Collapse
|
14
|
Roozitalab G, Abedi B, Imani S, Farghadani R, Jabbarzadeh Kaboli P. Comprehensive assessment of TECENTRIQ® and OPDIVO®: analyzing immunotherapy indications withdrawn in triple-negative breast cancer and hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:889-918. [PMID: 38409546 DOI: 10.1007/s10555-024-10174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 02/05/2024] [Indexed: 02/28/2024]
Abstract
Atezolizumab (TECENTRIQ®) and nivolumab (OPDIVO®) are both immunotherapeutic indications targeting programmed cell death 1 ligand 1 (PD-L1) and programmed cell death 1 (PD-1), respectively. These inhibitors hold promise as therapies for triple-negative breast cancer (TNBC) and hepatocellular carcinoma (HCC) and have demonstrated encouraging results in reducing the progression and spread of tumors. However, due to their adverse effects and low response rates, the US Food and Drug Administration (FDA) has withdrawn the approval of atezolizumab in TNBC and nivolumab in HCC treatment. The withdrawals of atezolizumab and nivolumab have raised concerns regarding their effectiveness and the ability to predict treatment responses. Therefore, the current study aims to investigate the immunotherapy withdrawal of PD-1/PD-L1 inhibitors, specifically atezolizumab for TNBC and nivolumab for HCC. This study will examine both the structural and clinical aspects. This review provides detailed insights into the structure of the PD-1 receptor and its ligands, the interactions between PD-1 and PD-L1, and their interactions with the withdrawn antibodies (atezolizumab and nivolumab) as well as PD-1 and PD-L1 modifications. In addition, this review further assesses these antibodies in the context of TNBC and HCC. It seeks to elucidate the factors that contribute to diverse responses to PD-1/PD-L1 therapy in different types of cancer and propose approaches for predicting responses, mitigating the potential risks linked to therapy withdrawals, and optimizing patient outcomes. By better understanding the mechanisms underlying responses to PD-1/PD-L1 therapy and developing strategies to predict these responses, it is possible to create more efficient treatments for TNBC and HCC.
Collapse
Affiliation(s)
- Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People's Republic of China
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan.
| |
Collapse
|
15
|
Hong Z, Liu F, Zhang Z. Ubiquitin modification in the regulation of tumor immunotherapy resistance mechanisms and potential therapeutic targets. Exp Hematol Oncol 2024; 13:91. [PMID: 39223632 PMCID: PMC11367865 DOI: 10.1186/s40164-024-00552-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Although immune checkpoint-based cancer immunotherapy has shown significant efficacy in various cancers, resistance still limits its therapeutic effects. Ubiquitination modification is a mechanism that adds different types of ubiquitin chains to proteins, mediating protein degradation or altering their function, thereby affecting cellular signal transduction. Increasing evidence suggests that ubiquitination modification plays a crucial role in regulating the mechanisms of resistance to cancer immunotherapy. Drugs targeting ubiquitination modification pathways have been shown to inhibit tumor progression or enhance the efficacy of cancer immunotherapy. This review elaborates on the mechanisms by which tumor cells, immune cells, and the tumor microenvironment mediate resistance to cancer immunotherapy and the details of how ubiquitination modification regulates these mechanisms, providing a foundation for enhancing the efficacy of cancer immunotherapy by intervening in ubiquitination modification.
Collapse
Affiliation(s)
- Zihang Hong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China
| | - Furong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
16
|
Tang J, Liu H, Li J, Zhang Y, Yao S, Yang K, You Z, Qiao X, Song Y. Regulation of post-translational modification of PD-L1 and associated opportunities for novel small-molecule therapeutics. Future Med Chem 2024; 16:1583-1599. [PMID: 38949857 PMCID: PMC11370925 DOI: 10.1080/17568919.2024.2366146] [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: 12/05/2023] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
PD-L1 is overexpressed on the surface of tumor cells and binds to PD-1, resulting in tumor immune escape. Therapeutic strategies to target the PD-1/PD-L1 pathway involve blocking the binding. Immune checkpoint inhibitors have limited efficacy against tumors because PD-L1 is also present in the cytoplasm. PD-L1 of post-translational modifications (PTMs) have uncovered numerous mechanisms contributing to carcinogenesis and have identified potential therapeutic targets. Therefore, small molecule inhibitors can block crucial carcinogenic signaling pathways, making them a potential therapeutic option. To better develop small molecule inhibitors, we have summarized the PTMs of PD-L1. This review discusses the regulatory mechanisms of small molecule inhibitors in carcinogenesis and explore their potential applications, proposing a novel approach for tumor immunotherapy based on PD-L1 PTM.
Collapse
Affiliation(s)
- Jinglin Tang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
| | - Han Liu
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
| | - Jinze Li
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
| | - Yibo Zhang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
| | - Suyang Yao
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
| | - Kan Yang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
- Key Laboratory of Medicinal Chemistry & Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei071002, China
| | - Zhihao You
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
| | - Xiaoqiang Qiao
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
- Key Laboratory of Medicinal Chemistry & Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei071002, China
| | - Yali Song
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei071002, China
- State Key Laboratory of New Pharmaceutical Preparations & Excipients, Hebei University, Baoding, Hebei071002, China
| |
Collapse
|
17
|
Chen G, Du D, Wang H, Li H. The Deubiquitinase USP22-Stabilized COL17A1 Promotes Lung Adenocarcinoma Progression. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13824. [PMID: 39143031 PMCID: PMC11324370 DOI: 10.1111/crj.13824] [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: 04/29/2024] [Revised: 06/19/2024] [Accepted: 07/26/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a highly aggressive and rapidly fatal malignancy worldwide. Collagen XVII (COL17A1) has been implicated in various protumorigenic processes. However, the functions and mechanisms of COL17A1 in LUAD progression still remain elusive. METHODS COL17A1 and ubiquitin-specific protease 22 (USP22) mRNA analysis was performed by quantitative PCR, and their protein levels were detected by immunoblotting and immunohistochemistry. The functional influence was evaluated by determining cell viability, proliferation, apoptosis, invasion, migration, and ferroptosis in vitro, as well as xenograft growth in vivo. Co-immunoprecipitation (Co-IP) and IP experiments were used to examine the USP22/COL17A1 interaction and COL17A1 deubiquitination. Cycloheximide treatment was used to analyze COL17A1 protein stability. RESULTS COL17A1 and USP22 were upregulated in human LUAD tissues and cell lines. Functionally, COL17A1 knockdown acted for the suppression of LUAD cell growth, invasion, and migration as well as promotion of cell apoptosis and ferroptosis in vitro. COL17A1 knockdown could diminish the tumorigenicity of LUAD cells in vivo. Mechanistically, USP22 stabilized and upregulated COL17A1 by enhancing the deubiquitination of COL17A1. Additionally, reexpression of COL17A1 could reverse USP22 silencing-induced phenotype changes of LUAD cells in vitro. CONCLUSION Our findings demonstrate that USP22-stabilized COL17A1 possesses oncogenic activity in LUAD. We propose that USP22 and COL17A1 would be potential targets for the establishment of therapeutic approaches against LUAD.
Collapse
Affiliation(s)
- Guangxi Chen
- Department of General Medicine, Jiujiang City Key Laboratory of Cell TherapyJiujiang NO.1 People's HospitalJiujiangChina
| | - Dandan Du
- Department of Jiulong Community Health Service Centre, Jiujiang City Key Laboratory of Cell TherapyJiujiang NO.1 People's HospitalJiujiangChina
| | - Haihua Wang
- Department of Chronic HepatologyJiujiang Third People's HospitalJiujiangChina
| | - Huifeng Li
- Department of Respiratory and Critical Care MedicineJiujiang Third People's HospitalJiujiangChina
| |
Collapse
|
18
|
Liu F, Chen J, Li K, Li H, Zhu Y, Zhai Y, Lu B, Fan Y, Liu Z, Chen X, Jia X, Dong Z, Liu K. Ubiquitination and deubiquitination in cancer: from mechanisms to novel therapeutic approaches. Mol Cancer 2024; 23:148. [PMID: 39048965 PMCID: PMC11270804 DOI: 10.1186/s12943-024-02046-3] [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: 04/17/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin-proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.
Collapse
Affiliation(s)
- Fangfang Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Jingyu Chen
- Department of Pediatric Medicine, School of Third Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Kai Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Haochen Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yiyi Zhu
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yubo Zhai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Bingbing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanle Fan
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Ziyue Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaojie Chen
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xuechao Jia
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Zigang Dong
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| | - Kangdong Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
19
|
Huang J, Yin Q, Wang Y, Zhou X, Guo Y, Tang Y, Cheng R, Yu X, Zhang J, Huang C, Huang Z, Zhang J, Guo Z, Huo X, Sun Y, Li Y, Wang H, Yang J, Xue L. EZH2 Inhibition Enhances PD-L1 Protein Stability Through USP22-Mediated Deubiquitination in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308045. [PMID: 38520088 PMCID: PMC11187912 DOI: 10.1002/advs.202308045] [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: 10/24/2023] [Revised: 02/26/2024] [Indexed: 03/25/2024]
Abstract
The regulation of PD-L1 is the key question, which largely determines the outcome of the immune checkpoint inhibitors (ICIs) based therapy. However, besides the transcription level, the protein stability of PD-L1 is closely correlated with its function and has drawn increasing attention. In this study, EZH2 inhibition enhances PD-L1 expression and protein stability, and the deubiquitinase ubiquitin-specific peptidase 22 (USP22) is identified as a key mediator in this process. EZH2 inhibition transcriptionally upregulates USP22 expression, and upregulated USP22 further stabilizes PD-L1. Importantly, a combination of EZH2 inhibitors with anti-PD-1 immune checkpoint blockade therapy improves the tumor microenvironment, enhances sensitivity to immunotherapy, and exerts synergistic anticancer effects. In addition, knocking down USP22 can potentially enhance the therapeutic efficacy of EZH2 inhibitors on colon cancer. These findings unveil the novel role of EZH2 inhibitors in tumor immune evasion by upregulating PD-L1, and this drawback can be compensated by combining ICI immunotherapy. Therefore, these findings provide valuable insights into the EZH2-USP22-PD-L1 regulatory axis, shedding light on the optimization of combining both immune checkpoint blockade and EZH2 inhibitor-based epigenetic therapies to achieve more efficacies and accuracy in cancer treatment.
Collapse
|
20
|
Cai M, Xu M, Yu D, Wang Q, Liu S. Posttranslational regulatory mechanism of PD-L1 in cancers and associated opportunities for novel small-molecule therapeutics. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1415-1424. [PMID: 38826132 PMCID: PMC11532205 DOI: 10.3724/abbs.2024085] [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: 02/26/2024] [Accepted: 04/03/2024] [Indexed: 06/04/2024] Open
Abstract
Despite the tremendous progress in cancer research over the past few decades, effective therapeutic strategies are still urgently needed. Accumulating evidence suggests that immune checkpoints are the cause of tumor immune escape. PD-1/PD-L1 are among them. Posttranslational modification is the most critical step for protein function, and the regulation of PD-L1 by small molecules through posttranslational modification is highly valuable. In this review, we discuss the mechanisms of tumor cell immune escape and several posttranslational modifications associated with PD-L1 and describe examples in which small molecules can regulate PD-L1 through posttranslational modifications. Herein, we propose that the use of small molecule compounds that act by inhibiting PD-L1 through posttranslational modifications is a promising therapeutic approach with the potential to improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Minchen Cai
- />Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Mengting Xu
- />Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Dianping Yu
- />Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Qun Wang
- />Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Sanhong Liu
- />Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| |
Collapse
|
21
|
Wang R, He S, Long J, Wang Y, Jiang X, Chen M, Wang J. Emerging therapeutic frontiers in cancer: insights into posttranslational modifications of PD-1/PD-L1 and regulatory pathways. Exp Hematol Oncol 2024; 13:46. [PMID: 38654302 DOI: 10.1186/s40164-024-00515-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024] Open
Abstract
The interaction between programmed cell death ligand 1 (PD-L1), which is expressed on the surface of tumor cells, and programmed cell death 1 (PD-1), which is expressed on T cells, impedes the effective activation of tumor antigen-specific T cells, resulting in the evasion of tumor cells from immune-mediated killing. Blocking the PD-1/PD-L1 signaling pathway has been shown to be effective in preventing tumor immune evasion. PD-1/PD-L1 blocking antibodies have garnered significant attention in recent years within the field of tumor treatments, given the aforementioned mechanism. Furthermore, clinical research has substantiated the efficacy and safety of this immunotherapy across various tumors, offering renewed optimism for patients. However, challenges persist in anti-PD-1/PD-L1 therapies, marked by limited indications and the emergence of drug resistance. Consequently, identifying additional regulatory pathways and molecules associated with PD-1/PD-L1 and implementing judicious combined treatments are imperative for addressing the intricacies of tumor immune mechanisms. This review briefly outlines the structure of the PD-1/PD-L1 molecule, emphasizing the posttranslational modification regulatory mechanisms and related targets. Additionally, a comprehensive overview on the clinical research landscape concerning PD-1/PD-L1 post-translational modifications combined with PD-1/PD-L1 blocking antibodies to enhance outcomes for a broader spectrum of patients is presented based on foundational research.
Collapse
Affiliation(s)
- Rong Wang
- Department of Pathology, Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Shiwei He
- School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China.
| | - Yian Wang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mingfen Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Fujian Medical University, Quanzhou, Fujian, China
| | - Jie Wang
- Department of Pathology, Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
22
|
Chai F, Li P, Liu X, Zhou Z, Ren H. Targeting the PD-L1 cytoplasmic domain and its regulatory pathways to enhance cancer immunotherapy. J Mol Cell Biol 2024; 15:mjad070. [PMID: 37993416 PMCID: PMC11193063 DOI: 10.1093/jmcb/mjad070] [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: 12/08/2022] [Revised: 05/09/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023] Open
Abstract
As a significant member of the immune checkpoint, programmed cell death 1 ligand 1 (PD-L1) plays a critical role in cancer immune escape and has become an important target for cancer immunotherapy. Clinically approved drugs mainly target the extracellular domain of PD-L1. Recently, the small cytoplasmic domain of PD-L1 has been reported to regulate PD-L1 stability and function through multiple pathways. Therefore, the intracellular domain of PD-L1 and its regulatory pathways could be promising targets for cancer therapy, expanding available strategies for combined immunotherapy. Here, we summarize the emerging roles of the PD-L1 cytoplasmic domain and its regulatory pathways. The conserved motifs, homodimerization, and posttranslational modifications of the PD-L1 cytoplasmic domain have been reported to regulate the membrane anchoring, degradation, nuclear translocation, and glycosylation of PD-L1. This summary provides a comprehensive understanding of the functions of the PD-L1 cytoplasmic domain and evaluates the broad prospects for targeted therapy.
Collapse
Affiliation(s)
- Fangni Chai
- Division of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Pan Li
- Division of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xin Liu
- Division of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zhihui Zhou
- Division of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Haiyan Ren
- Division of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
- Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| |
Collapse
|
23
|
Ren X, Wang L, Liu L, Liu J. PTMs of PD-1/PD-L1 and PROTACs application for improving cancer immunotherapy. Front Immunol 2024; 15:1392546. [PMID: 38638430 PMCID: PMC11024247 DOI: 10.3389/fimmu.2024.1392546] [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: 02/28/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Immunotherapy has been developed, which harnesses and enhances the innate powers of the immune system to fight disease, particularly cancer. PD-1 (programmed death-1) and PD-L1 (programmed death ligand-1) are key components in the regulation of the immune system, particularly in the context of cancer immunotherapy. PD-1 and PD-L1 are regulated by PTMs, including phosphorylation, ubiquitination, deubiquitination, acetylation, palmitoylation and glycosylation. PROTACs (Proteolysis Targeting Chimeras) are a type of new drug design technology. They are specifically engineered molecules that target specific proteins within a cell for degradation. PROTACs have been designed and demonstrated their inhibitory activity against the PD-1/PD-L1 pathway, and showed their ability to degrade PD-1/PD-L1 proteins. In this review, we describe how PROTACs target PD-1 and PD-L1 proteins to improve the efficacy of immunotherapy. PROTACs could be a novel strategy to combine with radiotherapy, chemotherapy and immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Xiaohui Ren
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lijuan Wang
- Department of Hospice Care, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Likun Liu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Liu
- Department of Special Needs Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
24
|
Zhang F, Jiang R, Sun S, Wu C, Yu Q, Awadasseid A, Wang J, Zhang W. Recent advances and mechanisms of action of PD-L1 degraders as potential therapeutic agents. Eur J Med Chem 2024; 268:116267. [PMID: 38422701 DOI: 10.1016/j.ejmech.2024.116267] [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: 09/07/2023] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
PD-L1 is an important immune checkpoint protein that can bind to T cells' PD-1 receptor, thereby promoting immune escape from tumors. In recent years, many researchers have developed strategies to degrade PD-L1 to improve the effect of immunotherapy. The study of degrading PD-L1 provides new opportunities for immunotherapy. Here, we mainly summarize and review the current active molecules and mechanisms that mediate the degradation of immature and mature PD-L1 during the post-translational modification stages, involving PD-L1 phosphorylation, glycosylation, palmitoylation, ubiquitination, and the autophagy-lysosomal process. This review expects that by degrading PD-L1 protein, we will not only gain a better understanding of oncogenic mechanisms involving tumor PD-L1 protein but also provide a new way to improve immunotherapy.
Collapse
Affiliation(s)
- Feng Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Ruiya Jiang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shishi Sun
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Caiyun Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Qimeng Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Annoor Awadasseid
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China; Moganshan Institute, Zhejiang University of Technology, Deqing, China
| | - Jianwei Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Wen Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
25
|
Kowald L, Roedig J, Karlowitz R, Wagner K, Smith S, Juretschke T, Beli P, Müller S, van Wijk SJL. USP22 regulates APL differentiation via PML-RARα stabilization and IFN repression. Cell Death Discov 2024; 10:128. [PMID: 38467608 PMCID: PMC10928094 DOI: 10.1038/s41420-024-01894-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/13/2024] Open
Abstract
Ubiquitin-specific peptidase 22 (USP22) is a deubiquitinating enzyme (DUB) that underlies tumorigenicity, proliferation, cell death and differentiation through deubiquitination of histone and non-histone targets. Ubiquitination determines stability, localization and functions of cell fate proteins and controls cell-protective signaling pathways to surveil cell cycle progression. In a variety of carcinomas, lymphomas and leukemias, ubiquitination regulates the tumor-suppressive functions of the promyelocytic leukemia protein (PML), but PML-specific DUBs, DUB-controlled PML ubiquitin sites and the functional consequences of PML (de)ubiquitination remain unclear. Here, we identify USP22 as regulator of PML and the oncogenic acute promyelocytic leukemia (APL) fusion PML-RARα protein stability and identify a destabilizing role of PML residue K394. Additionally, loss of USP22 upregulates interferon (IFN) and IFN-stimulated gene (ISG) expression in APL and induces PML-RARα stabilization and a potentiation of the cell-autonomous sensitivity towards all-trans retinoic acid (ATRA)-mediated differentiation. Our findings imply USP22-dependent surveillance of PML-RARα stability and IFN signaling as important regulator of APL pathogenesis, with implications for viral mimicry, differentiation and cell fate regulation in other leukemia subtypes.
Collapse
Affiliation(s)
- Lisa Kowald
- Institute for Experimental Pediatric Hematology and Oncology, Medical Faculty, Goethe-University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Jens Roedig
- Institute for Experimental Pediatric Hematology and Oncology, Medical Faculty, Goethe-University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Rebekka Karlowitz
- Institute for Experimental Pediatric Hematology and Oncology, Medical Faculty, Goethe-University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Kristina Wagner
- Institute of Biochemistry II (IBCII), Medical Faculty, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Sonja Smith
- Institute for Experimental Pediatric Hematology and Oncology, Medical Faculty, Goethe-University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Thomas Juretschke
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128, Mainz, Germany
| | - Petra Beli
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128, Mainz, Germany
| | - Stefan Müller
- Institute of Biochemistry II (IBCII), Medical Faculty, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Sjoerd J L van Wijk
- Institute for Experimental Pediatric Hematology and Oncology, Medical Faculty, Goethe-University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- University Cancer Centre Frankfurt (UCT), University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
26
|
Kim SB, Hwang S, Cha JY, Lee HJ. Programmed Death Ligand 1 Regulatory Crosstalk with Ubiquitination and Deubiquitination: Implications in Cancer Immunotherapy. Int J Mol Sci 2024; 25:2939. [PMID: 38474186 DOI: 10.3390/ijms25052939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Programmed death ligand 1 (PD-L1) plays a pivotal role in cancer immune evasion and is a critical target for cancer immunotherapy. This review focuses on the regulation of PD-L1 through the dynamic processes of ubiquitination and deubiquitination, which are crucial for its stability and function. Here, we explored the intricate mechanisms involving various E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) that modulate PD-L1 expression in cancer cells. Specific ligases are discussed in detail, highlighting their roles in tagging PD-L1 for degradation. Furthermore, we discuss the actions of DUBs that stabilize PD-L1 by removing ubiquitin chains. The interplay of these enzymes not only dictates PD-L1 levels but also influences cancer progression and patient response to immunotherapies. Furthermore, we discuss the therapeutic implications of targeting these regulatory pathways and propose novel strategies to enhance the efficacy of PD-L1/PD-1-based therapies. Our review underscores the complexity of PD-L1 regulation and its significant impact on the tumor microenvironment and immunotherapy outcomes.
Collapse
Affiliation(s)
- Soon-Bin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| | - Soonjae Hwang
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Ji-Young Cha
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Ho-Jae Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
27
|
Zhou Y, Wang F, Li G, Xu J, Zhang J, Gullen E, Yang J, Wang J. From immune checkpoints to therapies: understanding immune checkpoint regulation and the influence of natural products and traditional medicine on immune checkpoint and immunotherapy in lung cancer. Front Immunol 2024; 15:1340307. [PMID: 38426097 PMCID: PMC10902058 DOI: 10.3389/fimmu.2024.1340307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Lung cancer is a disease of global concern, and immunotherapy has brought lung cancer therapy to a new era. Besides promising effects in the clinical use of immune checkpoint inhibitors, immune-related adverse events (irAEs) and low response rates are problems unsolved. Natural products and traditional medicine with an immune-modulating nature have the property to influence immune checkpoint expression and can improve immunotherapy's effect with relatively low toxicity. This review summarizes currently approved immunotherapy and the current mechanisms known to regulate immune checkpoint expression in lung cancer. It lists natural products and traditional medicine capable of influencing immune checkpoints or synergizing with immunotherapy in lung cancer, exploring both their effects and underlying mechanisms. Future research on immune checkpoint modulation and immunotherapy combination applying natural products and traditional medicine will be based on a deeper understanding of their mechanisms regulating immune checkpoints. Continued exploration of natural products and traditional medicine holds the potential to enhance the efficacy and reduce the adverse reactions of immunotherapy.
Collapse
Affiliation(s)
- Yibin Zhou
- Department of Hematology and Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fenglan Wang
- Department of Hematology and Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Guangda Li
- Department of Hematology and Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Xu
- Department of Hematology and Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingjing Zhang
- Department of Hematology and Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Elizabeth Gullen
- Department of Pharmacology, Yale Medical School, New Haven, CT, United States
| | - Jie Yang
- Department of Hematology and Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Wang
- Department of Hematology and Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
28
|
Kuang X, Salinger A, Benavides F, Muller WJ, Dent SYR, Koutelou E. USP22 overexpression fails to augment tumor formation in MMTV-ERBB2 mice but loss of function impacts MMTV promoter activity. PLoS One 2024; 19:e0290837. [PMID: 38236941 PMCID: PMC10796002 DOI: 10.1371/journal.pone.0290837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/15/2023] [Indexed: 01/22/2024] Open
Abstract
The Ubiquitin Specific Peptidase 22 (USP22), a component of the Spt-Ada-Gcn5 Acetyltransferase (SAGA) histone modifying complex, is overexpressed in multiple human cancers, but how USP22 impacts tumorigenesis is not clear. We reported previously that Usp22 loss in mice impacts execution of several signaling pathways driven by growth factor receptors such as erythroblastic oncogene B b2 (ERBB2). To determine whether changes in USP22 expression affects ERBB2-driven tumorigenesis, we introduced conditional overexpression or deletion alleles of Usp22 into mice bearing the Mouse mammary tumor virus-Neu-Ires-Cre (MMTV-NIC) transgene, which drives both rat ERBB2/NEU expression and Cre recombinase activity from the MMTV promoter resulting in mammary tumor formation. We found that USP22 overexpression in mammary glands did not further enhance primary tumorigenesis in MMTV-NIC female mice, but increased lung metastases were observed. However, deletion of Usp22 significantly decreased tumor burden and increased survival of MMTV-NIC mice. These effects were associated with markedly decreased levels of both Erbb2 mRNA and protein, indicating Usp22 loss impacts MMTV promoter activity. Usp22 loss had no impact on ERBB2 expression in other settings, including MCF10A cells bearing a Cytomegalovirus (CMV)-driven ERBB2 transgene or in human epidermal growth factor receptor 2 (HER2)+ human SKBR3 and HCC1953 cells. Decreased activity of the MMTV promoter in MMTV-NIC mice correlated with decreased expression of known regulatory factors, including the glucocorticoid receptor (GR), the progesterone receptor (PR), and the chromatin remodeling factor Brahma-related gene-1 (BRG1). Together our findings indicate that increased expression of USP22 does not augment the activity of an activated ERBB2/NEU transgene but impacts of Usp22 loss on tumorigenesis cannot be assessed in this model due to unexpected effects on MMTV-driven Erbb2/Neu expression.
Collapse
Affiliation(s)
- Xianghong Kuang
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Andrew Salinger
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Fernando Benavides
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - William J. Muller
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Canada
- Department of Biochemistry, McGill University, Montreal, Canada
- Faculty of Medicine, McGill University, Montreal, Canada
| | - Sharon Y. R. Dent
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- The University of Texas MD Anderson Cancer Center/UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, United States of America
| | - Evangelia Koutelou
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
29
|
Ren W, Xu Z, Chang Y, Ju F, Wu H, Liang Z, Zhao M, Wang N, Lin Y, Xu C, Chen S, Rao Y, Lin C, Yang J, Liu P, Zhang J, Huang C, Xia N. Pharmaceutical targeting of OTUB2 sensitizes tumors to cytotoxic T cells via degradation of PD-L1. Nat Commun 2024; 15:9. [PMID: 38167274 PMCID: PMC10761827 DOI: 10.1038/s41467-023-44466-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
PD-1 is a co-inhibitory receptor expressed by CD8+ T cells which limits their cytotoxicity. PD-L1 expression on cancer cells contributes to immune evasion by cancers, thus, understanding the mechanisms that regulate PD-L1 protein levels in cancers is important. Here we identify tumor-cell-expressed otubain-2 (OTUB2) as a negative regulator of antitumor immunity, acting through the PD-1/PD-L1 axis in various human cancers. Mechanistically, OTUB2 directly interacts with PD-L1 to disrupt the ubiquitination and degradation of PD-L1 in the endoplasmic reticulum. Genetic deletion of OTUB2 markedly decreases the expression of PD-L1 proteins on the tumor cell surface, resulting in increased tumor cell sensitivity to CD8+ T-cell-mediated cytotoxicity. To underscore relevance in human patients, we observe a significant correlation between OTUB2 expression and PD-L1 abundance in human non-small cell lung cancer. An inhibitor of OTUB2, interfering with its deubiquitinase activity without disrupting the OTUB2-PD-L1 interaction, successfully reduces PD-L1 expression in tumor cells and suppressed tumor growth. Together, these results reveal the roles of OTUB2 in PD-L1 regulation and tumor evasion and lays down the proof of principle for OTUB2 targeting as therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Wenfeng Ren
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zilong Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yating Chang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
- School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Fei Ju
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hongning Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
- School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhiqi Liang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
- School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Min Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Naizhen Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yanhua Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chenhang Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
- School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shengming Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yipeng Rao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chaolong Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jianxin Yang
- Department of Hepatobiliary & Pancreatic Surgery, The National Key Clinical Specialty, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361004, China
- Fujian Provincial Key Laboratory and Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian, 361004, China
| | - Pingguo Liu
- Department of Hepatobiliary & Pancreatic Surgery, The National Key Clinical Specialty, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361004, China.
- Fujian Provincial Key Laboratory and Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian, 361004, China.
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Chenghao Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, Fujian, 361102, China.
- School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
30
|
Du Y, Zhang H, Hu H. Ubiquitination of Immune System and Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1466:35-45. [PMID: 39546134 DOI: 10.1007/978-981-97-7288-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Ubiquitination is a post-translational modification mechanism which regulates a variety of signaling pathways and crucial biological processes. It has long been known that ubiquitination regulates the fundamental cellular processes through the induction of proteasomal degradation of target proteins. Meanwhile, the nondegradative types of polyubiquitination modification have been appreciated as important regulatory machinery by modulating the activity or subcellular localization of key signaling proteins. The function of ubiquitination plays an important role in immune responses, which helps to maintain the stability of the internal environment and to control over protein stability and function and are thus critical for the regulation of both innate and adaptive immunity. Furthermore, ubiquitination also regulates both tumor-suppressing and tumor-promoting pathways in cancer. In this review, we will discuss recent progress regarding how ubiquitination regulates immune responses, focusing on Toll-like receptors signaling in innate immunity, T cell activation, TCR signaling, and tumor immunotherapy.
Collapse
Affiliation(s)
- Yizhou Du
- Center for Immunology and Hematology, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huiyuan Zhang
- Center for Immunology and Hematology, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Hu
- Center for Immunology and Hematology, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
31
|
Wang Y, Zhou Y, Yang L, Lei L, He B, Cao J, Gao H. Challenges Coexist with Opportunities: Spatial Heterogeneity Expression of PD-L1 in Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303175. [PMID: 37934012 PMCID: PMC10767451 DOI: 10.1002/advs.202303175] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/28/2023] [Indexed: 11/08/2023]
Abstract
Cancer immunotherapy using anti-programmed death-ligand 1 (PD-L1) antibodies has been used in various clinical applications and achieved certain results. However, such limitations as autoimmunity, tumor hyperprogression, and overall low patient response rate impede its further clinical application. Mounting evidence has revealed that PD-L1 is not only present in tumor cell membrane but also in cytoplasm, exosome, or even nucleus. Among these, the dynamic and spatial heterogeneous expression of PD-L1 in tumors is mainly responsible for the unsatisfactory efficacy of PD-L1 antibodies. Hence, numerous studies focus on inhibiting or degrading PD-L1 to improve immune response, while a comprehensive understanding of the molecular mechanisms underlying spatial heterogeneity of PD-L1 can fundamentally transform the current status of PD-L1 antibodies in clinical development. Herein, the concept of spatial heterogeneous expression of PD-L1 is creatively introduced, encompassing the structure and biological functions of various kinds of PD-L1 (including mPD-L1, cPD-L1, nPD-L1, and exoPD-L1). Then an in-depth analysis of the regulatory mechanisms and potential therapeutic targets of PD-L1 is provided, seeking to offer a solid basis for future investigation. Moreover, the current status of agents is summarized, especially small molecular modulators development directed at these new targets, offering a novel perspective on potential PD-L1 therapeutics strategies.
Collapse
Affiliation(s)
- Yazhen Wang
- National Engineering Research Center for BiomaterialsCollege of Biomedical EngineeringSichuan UniversityChengdu610064P. R. China
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education MinistrySichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041P. R. China
| | - Yang Zhou
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education MinistrySichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041P. R. China
| | - Lianyi Yang
- National Engineering Research Center for BiomaterialsCollege of Biomedical EngineeringSichuan UniversityChengdu610064P. R. China
| | - Lei Lei
- National Engineering Research Center for BiomaterialsCollege of Biomedical EngineeringSichuan UniversityChengdu610064P. R. China
| | - Bin He
- National Engineering Research Center for BiomaterialsCollege of Biomedical EngineeringSichuan UniversityChengdu610064P. R. China
| | - Jun Cao
- National Engineering Research Center for BiomaterialsCollege of Biomedical EngineeringSichuan UniversityChengdu610064P. R. China
| | - Huile Gao
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education MinistrySichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041P. R. China
| |
Collapse
|
32
|
Miao Z, Li J, Wang Y, Shi M, Gu X, Zhang X, Wei F, Tang X, Zheng L, Xing Y. Hsa_circ_0136666 stimulates gastric cancer progression and tumor immune escape by regulating the miR-375/PRKDC Axis and PD-L1 phosphorylation. Mol Cancer 2023; 22:205. [PMID: 38093288 PMCID: PMC10718020 DOI: 10.1186/s12943-023-01883-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/17/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Targeted drugs are not quite effective for prolonging the survival of patients with gastric cancer due to off-target effects as well as tumor immune escape mechanisms. Circular RNAs widely exist in tumor regions as biomarkers and can be developed as effective drug targets. METHODS Western blot, QRT-PCR, fluorescence in situ hybridization, and flow cytometry were used to investigate the function of hsa_circ_0136666 in promoting the proliferation of gastric cancer cells. Tissue immunofluorescence, enzyme-linked immunosorbent assay (ELISA), as well as flow cytometric analysis, was conducted to explore the process of tumor immune evasion in tumor-bearing mice. The differences of circRNA expression in clinical samples were analyzed through tissue microarray FISH. The effect of siRNA on improving the efficacy of anti-PDL1 drugs and suppressing the immune microenvironment was evaluated by the coadministration model. RESULTS We demonstrated that hsa_circ_0136666 was widely and highly expressed in gastric cancer tissues and cells. Functionally, hsa_circ_0136666 promoted gastric cancer tumor proliferation and tumor microenvironment formation, leading to tumorigenesis immune escape, and this effect was dependent on CD8 + T cells. Mechanistically, we confirmed that hsa_circ_0136666 competitively upregulated PRKDC expression by sponging miR-375-3p, regulating immune checkpoint proteins, prompting phosphorylation of PD-L1 to preventing its degradation, driving PD-L1 aggregation and suppressing immune function, thereby impairing cancer immune responses. In terms of application, we found that LNP-siRNA effectively improved anti-PDL1 drug efficacy and inhibited immune escape. CONCLUSION Our results reveal an oncogenic role played by hsa_circ_0136666 in gastric cancer, driving PD-L1 phosphorylation via the miR-375/PRKDC signaling axis, prompting immune escape. This work proposes a completely new pathogenic mechanism of gastric cancer, uncovers a novel role for hsa_circ_0136666 as an immune target, and provides a rationale for enhancing the efficacy of anti-PD-L1 therapy for gastric cancer.
Collapse
Affiliation(s)
- Zhenyan Miao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Jifei Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yu Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Mingqin Shi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xiao Gu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xuanqi Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Fang Wei
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xinying Tang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
33
|
Abstract
Ubiquitination is an essential regulator of most, if not all, signalling pathways, and defects in cellular signalling are central to cancer initiation, progression and, eventually, metastasis. The attachment of ubiquitin signals by E3 ubiquitin ligases is directly opposed by the action of approximately 100 deubiquitinating enzymes (DUBs) in humans. Together, DUBs and E3 ligases coordinate ubiquitin signalling by providing selectivity for different substrates and/or ubiquitin signals. The balance between ubiquitination and deubiquitination is exquisitely controlled to ensure properly coordinated proteostasis and response to cellular stimuli and stressors. Not surprisingly, then, DUBs have been associated with all hallmarks of cancer. These relationships are often complex and multifaceted, highlighted by the implication of multiple DUBs in certain hallmarks and by the impact of individual DUBs on multiple cancer-associated pathways, sometimes with contrasting cancer-promoting and cancer-inhibiting activities, depending on context and tumour type. Although it is still understudied, the ever-growing knowledge of DUB function in cancer physiology will eventually identify DUBs that warrant specific inhibition or activation, both of which are now feasible. An integrated appreciation of the physiological consequences of DUB modulation in relevant cancer models will eventually lead to the identification of patient populations that will most likely benefit from DUB-targeted therapies.
Collapse
Affiliation(s)
- Grant Dewson
- Ubiquitin Signalling Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
| | - Pieter J A Eichhorn
- Curtin Medical School, Curtin University, Perth, Western Australia, Australia.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| | - David Komander
- Ubiquitin Signalling Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
34
|
Ye Q, Zhou W, Xu S, Que Q, Zhan Q, Zhang L, Zheng S, Ling S, Xu X. Ubiquitin-specific protease 22 promotes tumorigenesis and progression by an FKBP12/mTORC1/autophagy positive feedback loop in hepatocellular carcinoma. MedComm (Beijing) 2023; 4:e439. [PMID: 38045832 PMCID: PMC10691294 DOI: 10.1002/mco2.439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 12/05/2023] Open
Abstract
Ubiquitin-specific protease 22 (USP22) has been identified as a potential marker for cancer stem cells in hepatocellular carcinoma (HCC). It can promote HCC stemness, which is considered a driver of tumorigenesis. Here, we sought to determine the role of USP22 in tumorigenesis, elucidate its underlying mechanism, and explore its therapeutic significance in HCC. As a result, we found that tissue-specific Usp22 overexpression accelerated tumorigenesis, whereas Usp22 ablation decelerated it in a c-Myc/NRasGV12-induced HCC mouse model and that the mammalian target of rapamycin complex 1 (mTORC1) pathway was activated downstream. USP22 overexpression resulted in increased tumorigenic properties that were reversed by rapamycin in vitro and in vivo. In addition, USP22 activated mTORC1 by deubiquitinating FK506-binding protein 12 (FKBP12) and activated mTORC1, in turn, further stabilizing USP22 by inhibiting autophagic degradation. Clinically, HCC patients with high USP22 expression tend to benefit from mTOR inhibitors after liver transplantation (LT). Our results revealed that USP22 promoted tumorigenesis and progression via an FKBP12/mTORC1/autophagy positive feedback loop in HCC. Clinically, USP22 may be an effective biomarker for selecting eligible recipients with HCC for anti-mTOR-based therapy after LT.
Collapse
Affiliation(s)
- Qianwei Ye
- Department of General SurgeryHangzhou First People's HospitalHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
- NHC Key Laboratory of Combined Multi‐Organ TransplantationHangzhouChina
| | - Wei Zhou
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Shengjun Xu
- Department of General SurgeryHangzhou First People's HospitalHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Qingyang Que
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Qifan Zhan
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Lincheng Zhang
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Shusen Zheng
- NHC Key Laboratory of Combined Multi‐Organ TransplantationHangzhouChina
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Sunbin Ling
- Department of General SurgeryHangzhou First People's HospitalHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Xiao Xu
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
35
|
Looi CK, Foong LC, Chung FFL, Khoo ASB, Loo EM, Leong CO, Mai CW. Targeting the crosstalk of epigenetic modifications and immune evasion in nasopharyngeal cancer. Cell Biol Toxicol 2023; 39:2501-2526. [PMID: 37755585 DOI: 10.1007/s10565-023-09830-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is a distinct type of head and neck cancer that is highly associated with Epstein-Barr virus (EBV) infection. EBV acts as an epigenetic driver in NPC tumorigenesis, reprogramming the viral and host epigenomes to regulate viral latent gene expression, and creating an environment conducive to the malignant transformation of nasopharyngeal epithelial cells. Targeting epigenetic mechanisms in pre-clinical studies has been shown promise in eradicating tumours and overcoming immune resistance in some solid tumours. However, its efficacy in NPC remains inclusive due to the complex nature of this cancer. In this review, we provide an updated understanding of the roles of epigenetic factors in regulating EBV latent gene expression and promoting NPC progression. We also explore the crosstalk between epigenetic mechanisms and immune evasion in NPC. Particularly, we discuss the potential roles of DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors in reversing immune suppression and augmenting antitumour immunity. Furthermore, we highlight the advantages of combining epigenetic therapy and immune checkpoint inhibitor to reverse immune resistance and improve clinical outcomes. Epigenetic drugs have the potential to modulate both epigenetic mediators and immune factors involved in NPC. However, further research is needed to fully comprehend the diverse range of epigenetic modifications in NPC. A deeper understanding of the crosstalk between epigenetic mechanisms and immune evasion during NPC progression is crucial for the development of more effective treatments for this challenging disease.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Studies, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Lian-Chee Foong
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Pudong New District, Shanghai, 200127, China
| | - Felicia Fei-Lei Chung
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Alan Soo-Beng Khoo
- School of Postgraduate Studies, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Pennsylvania, PA, 19107, USA
| | - Ee-Mun Loo
- AGTC Genomics, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, UCSI Heights, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Chee-Onn Leong
- AGTC Genomics, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, Development, and Innovation (IRDI), Institute for Research, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Chun-Wai Mai
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Pudong New District, Shanghai, 200127, China.
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, UCSI Heights, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
36
|
Zhang Y, Song J, Zhou Y, Jia H, Zhou T, Sun Y, Gao Q, Zhao Y, Pan Y, Sun Z, Chu P. Discovery of selective and potent USP22 inhibitors via structure-based virtual screening and bioassays exerting anti-tumor activity. Bioorg Chem 2023; 141:106842. [PMID: 37769523 DOI: 10.1016/j.bioorg.2023.106842] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/20/2023] [Accepted: 09/05/2023] [Indexed: 10/03/2023]
Abstract
Ubiquitin-specific protease 22 (USP22) plays a prominent role in tumor development, invasion, metastasis and immune reprogramming, which has been proposed as a potential therapeutic target for cancer. Herein, we employed a structure-based discovery and biological evaluation and discovered that Rottlerin (IC50 = 2.53 μM) and Morusin (IC50 = 8.29 μM) and as selective and potent USP22 inhibitors. Treatment of HCT116 cells and A375 cells with each of the two compounds resulted in increased monoubiquitination of histones H2A and H2B, as well as reduced protein expression levels of Sirt1 and PD-L1, all of which are known as USP22 substrates. Additionally, our study demonstrated that the administration of Rottlerin or Morusin resulted in an increase H2Bub levels, while simultaneously reducing the expression of Sirt1 and PD-L1 in a manner dependent on USP22. Furthermore, Rottlerin and Morusin were found to enhance the degradation of PD-L1 and Sirt1, as well as increase the polyubiquitination of endogenous PD-L1 and Sirt1 in HCT116 cells. Moreover, in an in vivo syngeneic tumor model, Rottlerin and Morusin exhibited potent antitumor activity, which was accompanied by an enhanced infiltration of T cells into the tumor tissues. Using in-depth molecular dynamics (MD) and binding free energy calculation, conserved residue Leu475 and non-conserved residue Arg419 were proven to be crucial for the binding affinity and inhibitory function of USP22 inhibitors. In summary, our study established a highly efficient approach for USP22-specific inhibitor discovery, which lead to identification of two selective and potent USP22 inhibitors as potential drugs in anticancer therapy.
Collapse
Affiliation(s)
- Yue Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Jiankun Song
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China
| | - Yuanzhang Zhou
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China
| | - Huijun Jia
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China
| | - Tianyu Zhou
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yingbo Sun
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Qiong Gao
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China
| | - Yue Zhao
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China
| | - Yujie Pan
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Zhaolin Sun
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China.
| | - Peng Chu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
37
|
Zhang K, Sun T, Li W, Guo Y, Li A, Hsieh M, Wang J, Wu J, Arvanitis L, Raz DJ. Inhibition of USP7 upregulates USP22 and activates its downstream cancer-related signaling pathways in human cancer cells. Cell Commun Signal 2023; 21:319. [PMID: 37946202 PMCID: PMC10634000 DOI: 10.1186/s12964-023-01320-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/14/2023] [Indexed: 11/12/2023] Open
Abstract
Deubiquitinases (DUBs) play important roles in various human cancers and targeting DUBs is considered as a novel anticancer therapeutic strategy. Overexpression of ubiquitin specific protease 7 and 22 (USP7 and USP22) are associated with malignancy, therapy resistance, and poor prognosis in many cancers. Although both DUBs are involved in the regulation of similar genes and signaling pathways, such as histone H2B monoubiquitination (H2Bub1), c-Myc, FOXP3, and p53, the interdependence of USP22 and USP7 expression has never been described. In the study, we found that targeting USP7 via either siRNA-mediated knockdown or pharmaceutical inhibitors dramatically upregulates USP22 in cancer cells. Mechanistically, the elevated USP22 occurs through a transcriptional pathway, possibly due to desuppression of the transcriptional activity of SP1 via promoting its degradation upon USP7 inhibition. Importantly, increased USP22 expression leads to significant activation of downstream signal pathways including H2Bub1 and c-Myc, which may potentially enhance cancer malignancy and counteract the anticancer efficacy of USP7 inhibition. Importantly, targeting USP7 further suppresses the in vitro proliferation of USP22-knockout (USP22-Ko) A549 and H1299 lung cancer cells and induces a stronger activation of p53 tumor suppressor signaling pathway. In addition, USP22-Ko cancer cells are more sensitive to a combination of cisplatin and USP7 inhibitor. USP7 inhibitor treatment further suppresses in vivo angiogenesis and tumor growth and induced more apoptosis in USP22-Ko cancer xenografts. Taken together, our findings demonstrate that USP7 inhibition can dramatically upregulate USP22 in cancer cells; and targeting USP7 and USP22 may represent a more effective approach for targeted cancer therapy, which warrants further study. Video Abstract.
Collapse
Affiliation(s)
- Keqiang Zhang
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA.
| | - Ting Sun
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA
- Faculty of Health Science, University of Macau, Macau, China
| | - Wendong Li
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Yuming Guo
- Division of Comparative Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Aimin Li
- Pathology Core of Shared Resources, City of Hope National Medical Center, Duarte, CA, USA
| | - Marcus Hsieh
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Jinghan Wang
- Department of Hepatobiliary and Pancreatic Surgery, East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Wu
- Division of Comparative Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Leonidas Arvanitis
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Dan J Raz
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
38
|
Friebus-Kardash J, Christ TC, Dietlein N, Elwy A, Abdelrahman H, Holnsteiner L, Hu Z, Rodewald HR, Lang KS. Usp22 Deficiency Leads to Downregulation of PD-L1 and Pathological Activation of CD8 + T Cells and Causes Immunopathology in Response to Acute LCMV Infection. Vaccines (Basel) 2023; 11:1563. [PMID: 37896966 PMCID: PMC10610587 DOI: 10.3390/vaccines11101563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/07/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Ubiquitin-specific peptidase 22 (Usp22) cleaves ubiquitin moieties from numerous proteins, including histone H2B and transcription factors. Recently, it was reported that Usp22 acts as a negative regulator of interferon-dependent responses. In the current study, we investigated the role of Usp22 deficiency in acute viral infection with lymphocytic choriomeningitis virus (LCMV). We found that the lack of Usp22 on bone marrow-derived cells (Usp22fl/fl Vav1-Cre mice) reduced the induction of type I and II interferons. A limited type I interferon response did not influence virus replication. However, restricted expression of PD-L1 led to increased frequencies of functional virus-specific CD8+ T cells and rapid death of Usp22-deficient mice. CD8+ T cell depletion experiments revealed that accelerated CD8+ T cells were responsible for enhanced lethality in Usp22 deficient mice. In conclusion, we found that the lack of Usp22 generated a pathological CD8+ T cell response, which gave rise to severe disease in mice.
Collapse
Affiliation(s)
- Justa Friebus-Kardash
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Theresa Charlotte Christ
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Nikolaus Dietlein
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; (N.D.)
| | - Abdelrahman Elwy
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Hossam Abdelrahman
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Lisa Holnsteiner
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Zhongwen Hu
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; (N.D.)
| | - Karl Sebastian Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| |
Collapse
|
39
|
Mu N, Wang Y, Li X, Du Z, Wu Y, Su M, Wang Y, Sun X, Su L, Liu X. Crotonylated BEX2 interacts with NDP52 and enhances mitophagy to modulate chemotherapeutic agent-induced apoptosis in non-small-cell lung cancer cells. Cell Death Dis 2023; 14:645. [PMID: 37777549 PMCID: PMC10542755 DOI: 10.1038/s41419-023-06164-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 10/02/2023]
Abstract
Brain expressed X-linked gene 2 (BEX2) encoded protein was originally identified to promote transcription by interacting with several transcription factors in the DNA-binding complexes. Recently, BEX2 was found to be localized in cytosol and/or mitochondria and regulate apoptosis in cancer cells and tumor growth. However, the molecular mechanism underlying its roles in cancer cells remains unclear. Here, we report that crotonylated BEX2 plays an important role in inhibiting chemotherapeutic agent-induced apoptosis via enhancing mitophagy in human lung cancer cells. BEX2 promotes mitophagy by facilitating interaction between NDP52 and LC3B. Moreover, BEX2 crotonylation at K59 is critical in the BEX2-mediated mitophagy in lung cancer cells. The K59R mutation of BEX2 inhibits mitophagy by affecting the interaction of NDP52 and LC3B. BEX2 expression is elevated after anticancer drug treatment, and its overexpression inhibits chemotherapy-induced apoptosis. In addition, inhibition of BEX2-regulated mitophagy sensitizes tumor cells to apoptosis. Furthermore, BEX2 promotes tumor growth and inhibits apoptosis by regulating mitophagy in vivo. We also confirm that BEX2 is overexpressed in lung adenocarcinoma and is associated with poor prognosis in lymph node metastasis-free cancer. Therefore, combination treatment with pharmaceutical approaches targeting BEX2-induced mitophagy and anticancer drugs may represent a potential strategy for NSCLC therapy.
Collapse
Affiliation(s)
- Ning Mu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
- The Second Hospital, Shandong University, Jinan, China
| | - Yu Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
- Qilu Hospital, Shandong University, Jinan, China
| | - Xiaopeng Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Zhiyuan Du
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yingdi Wu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Min Su
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yingying Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiaoyang Sun
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Ling Su
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| | - Xiangguo Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| |
Collapse
|
40
|
Feng C, Zhang L, Chang X, Qin D, Zhang T. Regulation of post-translational modification of PD-L1 and advances in tumor immunotherapy. Front Immunol 2023; 14:1230135. [PMID: 37554324 PMCID: PMC10405826 DOI: 10.3389/fimmu.2023.1230135] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/10/2023] [Indexed: 08/10/2023] Open
Abstract
The immune checkpoint molecules programmed cell death receptor 1 (PD-1) and programmed death ligand 1 (PD-L1) are one of the most promising targets for tumor immunotherapy. PD-L1 is overexpressed on the surface of tumor cells and inhibits T cell activation upon binding to PD⁃1 on the surface of T cells, resulting in tumor immune escape. The therapeutic strategy of targeting PD-1/PD-L1 involves blocking this binding and restoring the tumor-killing effect of immune cells. However, in clinical settings, a relatively low proportion of cancer patients have responded well to PD-1/PD-L1 blockade, and clinical outcomes have reached a bottleneck and no substantial progress has been made. In recent years, PD-L1 post-translation modifications (PTMs) have gradually become a hot topic in the field of PD-L1 research, which will provide new insights to improve the efficacy of current anti-PD-1/PD-L1 therapies. Here, we summarized and discussed multiple PTMs of PD-L1, including glycosylation, ubiquitination, phosphorylation, acetylation and palmitoylation, with a major emphasis on mechanism-based therapeutic strategies (including relevant enzymes and targets that are already in clinical use and that may become drugs in the future). We also summarized the latest research progress of PTMs of PD-L1/PD-1 in regulating immunotherapy. The review provided novel strategies and directions for tumor immunotherapy research based on the PTMs of PD-L1/PD-1.
Collapse
Affiliation(s)
- Chong Feng
- Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Lening Zhang
- Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xin Chang
- Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Dongliang Qin
- Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Tao Zhang
- Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
41
|
Wang M, Zhang Z, Li Z, Zhu Y, Xu C. E3 ubiquitin ligases and deubiquitinases in bladder cancer tumorigenesis and implications for immunotherapies. Front Immunol 2023; 14:1226057. [PMID: 37497216 PMCID: PMC10366618 DOI: 10.3389/fimmu.2023.1226057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 06/23/2023] [Indexed: 07/28/2023] Open
Abstract
With the rapidly increasing incidence of bladder cancer in China and worldwide, great efforts have been made to understand the detailed mechanism of bladder cancer tumorigenesis. Recently, the introduction of immune checkpoint inhibitor-based immunotherapy has changed the treatment strategy for bladder cancer, especially for advanced bladder cancer, and has improved the survival of patients. The ubiquitin-proteasome system, which affects many biological processes, plays an important role in bladder cancer. Several E3 ubiquitin ligases and deubiquitinases target immune checkpoints, either directly or indirectly. In this review, we summarize the recent progress in E3 ubiquitin ligases and deubiquitinases in bladder cancer tumorigenesis and further highlight the implications for bladder cancer immunotherapies.
Collapse
Affiliation(s)
- Maoyu Wang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhensheng Zhang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhizhou Li
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yasheng Zhu
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chuanliang Xu
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
42
|
Yang Y, Song R, Gao Y, Yu H, Wang S. Regulatory mechanisms and therapeutic potential of JAB1 in neurological development and disorders. Mol Med 2023; 29:80. [PMID: 37365502 DOI: 10.1186/s10020-023-00675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
c-Jun activation domain binding protein-1 (JAB1) is a multifunctional regulator that plays vital roles in diverse cellular processes. It regulates AP-1 transcriptional activity and also acts as the fifth component of the COP9 signalosome complex. While JAB1 is considered an oncoprotein that triggers tumor development, recent studies have shown that it also functions in neurological development and disorders. In this review, we summarize the general features of the JAB1 gene and protein, and present recent updates on the regulation of JAB1 expression. Moreover, we also highlight the functional roles and regulatory mechanisms of JAB1 in neurodevelopmental processes such as neuronal differentiation, synaptic morphogenesis, myelination, and hair cell development and in the pathogenesis of some neurological disorders such as Alzheimer's disease, multiple sclerosis, neuropathic pain, and peripheral nerve injury. Furthermore, current challenges and prospects are discussed, including updates on drug development targeting JAB1.
Collapse
Affiliation(s)
- Yu Yang
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Ruying Song
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Yiming Gao
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Hao Yu
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China.
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China.
| | - Shuai Wang
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China.
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
43
|
Jiang Y, Hong K, Zhao Y, Xu K. Emerging role of deubiquitination modifications of programmed death-ligand 1 in cancer immunotherapy. Front Immunol 2023; 14:1228200. [PMID: 37415977 PMCID: PMC10321661 DOI: 10.3389/fimmu.2023.1228200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 07/08/2023] Open
Abstract
Immune evasion is essential for carcinogenesis and cancer progression. Programmed death-ligand 1 (PD-L1), a critical immune checkpoint molecule, interacts with programmed death receptor-1 (PD-1) on immune cells to suppress anti-tumor immune responses. In the past decade, antibodies targeting PD-1/PD-L1 have tremendously altered cancer treatment paradigms. Post-translational modifications have been reported as key regulators of PD-L1 expression. Among these modifications, ubiquitination and deubiquitination are reversible processes that dynamically control protein degradation and stabilization. Deubiquitinating enzymes (DUBs) are responsible for deubiquitination and have emerged as crucial players in tumor growth, progression, and immune evasion. Recently, studies have highlighted the participation of DUBs in deubiquitinating PD-L1 and modulating its expression. Here, we review the recent developments in deubiquitination modifications of PD-L1 and focus on the underlying mechanisms and effects on anti-tumor immunity.
Collapse
Affiliation(s)
- Yao Jiang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Hong
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingchao Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Wang X, Zhang Y, Wu Y, Cheng H, Wang X. The role of E3 ubiquitin ligases and deubiquitinases in bladder cancer development and immunotherapy. Front Immunol 2023; 14:1202633. [PMID: 37215134 PMCID: PMC10196180 DOI: 10.3389/fimmu.2023.1202633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Bladder cancer is one of the common malignant urothelial tumors. Post-translational modification (PTMs), including ubiquitination, acetylation, methylation, and phosphorylation, have been revealed to participate in bladder cancer initiation and progression. Ubiquitination is the common PTM, which is conducted by E1 ubiquitin-activating enzyme, E2 ubiquitin-conjugating enzyme and E3 ubiquitin-protein ligase. E3 ubiquitin ligases play a key role in bladder oncogenesis and progression and drug resistance in bladder cancer. Therefore, in this review, we summarize current knowledge regarding the functions of E3 ubiquitin ligases in bladder cancer development. Moreover, we provide the evidence of E3 ubiquitin ligases in regulation of immunotherapy in bladder cancer. Furthermore, we mention the multiple compounds that target E3 ubiquitin ligases to improve the therapy efficacy of bladder cancer. We hope our review can stimulate researchers and clinicians to investigate whether and how targeting E3 ubiquitin ligases acts a novel strategy for bladder cancer therapy.
Collapse
|
45
|
Zhang Y, Zhang X, Li F, Lin C, Zhang D, Duan B, Zhao Y, Li X, Xu D, Cheng J, Zhao L, Wang J, Wang W. Expression profiles of the CD274 and PLEKHH2 gene and association of its polymorphism with hematologic parameters in sheep. Vet Immunol Immunopathol 2023; 259:110597. [PMID: 37094535 DOI: 10.1016/j.vetimm.2023.110597] [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: 11/28/2022] [Revised: 04/10/2023] [Accepted: 04/20/2023] [Indexed: 04/26/2023]
Abstract
CD274 and PLEKHH2 genes have been identified as immune- and multiple diseases-related genes, and have recently garnered significant interest. However, their role in regulating immune functions in sheep remains largely unexplored. In this study, we aimed to investigate the effects of polymorphisms in CD274 and PLEKHH2 on hematologic parameters in 915 sheep. Our results showed that the CD274 and PLEKHH2 genes were most highly expressed in the spleen and tail fat, respectively, as determined by qRT-PCR. We also identified a G to A mutation (g 0.11858 G > A) in the exon 4 region of CD274, and a C to G mutation (g 0.38384 C > G) in the intron 8 region of PLEKH2. Association analysis revealed that CD274 g 0.11858 G > A was significantly associated with RBC, HCT, MCHC, and MCV (P < 0.05), while PLEKHH2 g 0.38384 C > G was significantly associated with HCT, MPV, MCHC, and MCV (P < 0.05). These results suggest that CD274 and PLEKHH2 genes may play a role in regulating blood physiological indicators and could be potential functional candidates for influencing immune traits in sheep breeding programs.
Collapse
Affiliation(s)
- Yukun Zhang
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China; State Key Laboratory of Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, Lanzhou University, Lanzhou 730020, China
| | - Xiaoxue Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Fadi Li
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China; State Key Laboratory of Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, Lanzhou University, Lanzhou 730020, China
| | - Changchun Lin
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Deyin Zhang
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China; State Key Laboratory of Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, Lanzhou University, Lanzhou 730020, China
| | - Benzhen Duan
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200433, China; Key Laboratory of Medical Molecular Virology, MOE & NHC, School of Basic Medical Sciences, Fudan University, Shanghai 200433, China
| | - Yuan Zhao
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China; State Key Laboratory of Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, Lanzhou University, Lanzhou 730020, China
| | - Xiaolong Li
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China; State Key Laboratory of Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, Lanzhou University, Lanzhou 730020, China
| | - Dan Xu
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China; State Key Laboratory of Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, Lanzhou University, Lanzhou 730020, China
| | - Jiangbo Cheng
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China; State Key Laboratory of Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, Lanzhou University, Lanzhou 730020, China
| | - Liming Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jianghui Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Weimin Wang
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China; State Key Laboratory of Grassland Agro-Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Engineering Research Center of Grassland Industry, Ministry of Education, Lanzhou University, Lanzhou 730020, China.
| |
Collapse
|
46
|
Wang H, Langlais D, Nijnik A. Histone H2A deubiquitinases in the transcriptional programs of development and hematopoiesis: a consolidated analysis. Int J Biochem Cell Biol 2023; 157:106384. [PMID: 36738766 DOI: 10.1016/j.biocel.2023.106384] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Monoubiquitinated lysine 119 of histone H2A (H2AK119ub) is a highly abundant epigenetic mark, associated with gene repression and deposited on chromatin by the polycomb repressor complex 1 (PRC1), which is an essential regulator of diverse transcriptional programs in mammalian development and tissue homeostasis. While multiple deubiquitinases (DUBs) with catalytic activity for H2AK119ub (H2A-DUBs) have been identified, we lack systematic analyses of their roles and cross-talk in transcriptional regulation. Here, we address H2A-DUB functions in epigenetic regulation of mammalian development and tissue maintenance by conducting a meta-analysis of 248 genomics datasets from 32 independent studies, focusing on the mouse model and covering embryonic stem cells (ESCs), hematopoietic, and immune cell lineages. This covers all the publicly available datasets that map genomic H2A-DUB binding and H2AK119ub distributions (ChIP-Seq), and all datasets assessing dysregulation in gene expression in the relevant H2A-DUB knockout models (RNA-Seq). Many accessory datasets for PRC1-2 and DUB-interacting proteins are also analyzed and interpreted, as well as further data assessing chromatin accessibility (ATAC-Seq) and transcriptional activity (RNA-seq). We report co-localization in the binding of H2A-DUBs BAP1, USP16, and to a lesser extent others that is conserved across different cell-types, and also the enrichment of antagonistic PRC1-2 protein complexes at the same genomic locations. Such conserved sites enriched for the H2A-DUBs and PRC1-2 are proximal to transcriptionally active genes that engage in housekeeping cellular functions. Nevertheless, they exhibit H2AK119ub levels significantly above the genomic average that can undergo further increase with H2A-DUB knockout. This indicates a cooperation between H2A-DUBs and PRC1-2 in the modulation of housekeeping transcriptional programs, conserved across many cell types, likely operating through their antagonistic effects on H2AK119ub and the regulation of local H2AK119ub turnover. Our study further highlights existing knowledge gaps and discusses important directions for future work.
Collapse
Affiliation(s)
- HanChen Wang
- Department of Physiology, McGill University, Montreal, QC, Canada; McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - David Langlais
- McGill University Research Centre on Complex Traits, McGill University, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada; McGill Genome Centre, Montreal, QC, Canada.
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, QC, Canada; McGill University Research Centre on Complex Traits, McGill University, QC, Canada.
| |
Collapse
|
47
|
Hou B, Chen T, Zhang H, Li J, Wang P, Shang G. The E3 ubiquitin ligases regulate PD-1/PD-L1 protein levels in tumor microenvironment to improve immunotherapy. Front Immunol 2023; 14:1123244. [PMID: 36733484 PMCID: PMC9887025 DOI: 10.3389/fimmu.2023.1123244] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
The tumor microenvironment (TME) is the tumor surrounding environment, which is critical for tumor development and progression. TME is also involved in clinical intervention and treatment outcomes. Modulation of TME is useful for improving therapy strategies. PD-L1 protein on tumor cells interacts with PD-1 protein on T cells, contributing to T cell dysfunction and exhaustion, blockage of the immune response. Evidence has demonstrated that the expression of PD-1/PD-L1 is associated with clinical response to anti-PD-1/PD-L1 therapy in cancer patients. It is important to discuss the regulatory machinery how PD-1/PD-L1 protein is finely regulated in tumor cells. In recent years, studies have demonstrated that PD-1/PD-L1 expression was governed by various E3 ubiquitin ligases in TME, contributing to resistance of anti-PD-1/PD-L1 therapy in human cancers. In this review, we will discuss the role and molecular mechanisms of E3 ligases-mediated regulation of PD-1 and PD-L1 in TME. Moreover, we will describe how E3 ligases-involved PD-1/PD-L1 regulation alters anti-PD-1/PD-L1 efficacy. Altogether, targeting E3 ubiquitin ligases to control the PD-1/PD-L1 protein levels could be a potential strategy to potentiate immunotherapeutic effects in cancer patients.
Collapse
Affiliation(s)
- Bo Hou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiatong Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peter Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, China
| | - Guanning Shang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China,*Correspondence: Guanning Shang,
| |
Collapse
|
48
|
Roshani M, Baniebrahimi G, Mousavi M, Zare N, Sadeghi R, Salarinia R, Sheida A, Molavizadeh D, Sadeghi S, Moammer F, Zolfaghari MR, Mirzaei H. Exosomal long non-coding RNAs: novel molecules in gastrointestinal cancers' progression and diagnosis. Front Oncol 2022; 12:1014949. [PMID: 36591473 PMCID: PMC9795196 DOI: 10.3389/fonc.2022.1014949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal (GI) cancers arise in the GI tract and accessory organs, including the mouth, esophagus, stomach, liver, biliary tract, pancreas, small intestine, large intestine, and rectum. GI cancers are a major cause of cancer-related morbidity and mortality worldwide. Exosomes act as mediators of cell-to-cell communication, with pleiotropic activity in the regulation of homeostasis, and can be markers for diseases. Non-coding RNAs (ncRNAs), such as long non-coding RNAs (lncRNAs), can be transported by exosomes derived from tumor cells or non-tumor cells. They can be taken by recipient cells to alter their function or remodel the tumor microenvironment. Moreover, due to their uniquely low immunogenicity and excellent stability, exosomes can be used as natural carriers for therapeutic ncRNAs in vivo. Exosomal lncRNAs have a crucial role in regulating several cancer processes, including angiogenesis, proliferation, drug resistance, metastasis, and immunomodulation. Exosomal lncRNA levels frequently alter according to the onset and progression of cancer. Exosomal lncRNAs can therefore be employed as biomarkers for the diagnosis and prognosis of cancer. Exosomal lncRNAs can also monitor the patient's response to chemotherapy while also serving as potential targets for cancer treatment. Here, we discuss the role of exosomal lncRNAs in the biology and possible future treatment of GI cancer.
Collapse
Affiliation(s)
- Mohammad Roshani
- Internal Medicine and Gastroenterology, Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Baniebrahimi
- Department of Pediatric Dentistry, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Mousavi
- Department of Anatomy, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Noushid Zare
- Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Reza Sadeghi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Salarinia
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Sciences, Bojnurd, Iran
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Danial Molavizadeh
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Sadeghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Farzaneh Moammer
- Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Hamed Mirzaei
- Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
49
|
Li Q, Zhang L, You W, Xu J, Dai J, Hua D, Zhang R, Yao F, Zhou S, Huang W, Dai Y, Zhang Y, Baheti T, Qian X, Pu L, Xu J, Xia Y, Zhang C, Tang J, Wang X. PRDM1/BLIMP1 induces cancer immune evasion by modulating the USP22-SPI1-PD-L1 axis in hepatocellular carcinoma cells. Nat Commun 2022; 13:7677. [PMID: 36509766 PMCID: PMC9744896 DOI: 10.1038/s41467-022-35469-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Programmed death receptor-1 (PD-1) blockade have achieved some efficacy but only in a fraction of patients with hepatocellular carcinoma (HCC). Programmed cell death 1 ligand 1 (PD-L1) binds to its receptor PD1 on T cells to dampen antigen-tumor immune responses. However, the mechanisms underlying PD-L1 regulation are not fully elucidated. Herein, we identify that tumoral Prdm1 overexpression inhibits cell growth in immune-deficient mouse models. Further, tumoral Prdm1 overexpression upregulates PD-L1 levels, dampening anti-tumor immunity in vivo, and neutralizes the anti-tumor efficacy of Prdm1 overexpression in immune-competent mouse models. Mechanistically, PRDM1 enhances USP22 transcription, thus reducing SPI1 protein degradation through deubiquitination, which enhances PD-L1 transcription. Functionally, PD-1 mAb treatment reinforces the efficacy of Prdm1-overexpressing HCC immune-competent mouse models. Collectively, we demonstrate that the PRDM1-USP22-SPI1 axis regulates PD-L1 levels, resulting in infiltrated CD8+ T cell exhaustion. Furthermore, PRDM1 overexpression combined with PD-(L)1 mAb treatment provides a therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Qing Li
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Liren Zhang
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Wenhua You
- grid.263826.b0000 0004 1761 0489School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu Province China ,grid.89957.3a0000 0000 9255 8984Department of Immunology, Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, Jiangsu Province China
| | - Jiali Xu
- grid.412676.00000 0004 1799 0784Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province China
| | - Jingjing Dai
- grid.412676.00000 0004 1799 0784Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province China
| | - Dongxu Hua
- grid.89957.3a0000 0000 9255 8984The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Ruizhi Zhang
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Feifan Yao
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Suiqing Zhou
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Wei Huang
- grid.411610.30000 0004 1764 2878Department of General Surgery, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Ili, China
| | - Yongjiu Dai
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Yu Zhang
- grid.411610.30000 0004 1764 2878Department of General Surgery, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Ili, China
| | - Tasiken Baheti
- grid.411610.30000 0004 1764 2878Department of General Surgery, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Ili, China
| | - Xiaofeng Qian
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Liyong Pu
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Jing Xu
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province China
| | - Yongxiang Xia
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Chuanyong Zhang
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| | - Jinhai Tang
- grid.412676.00000 0004 1799 0784Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province China
| | - Xuehao Wang
- grid.89957.3a0000 0000 9255 8984Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province China
| |
Collapse
|
50
|
Mao M, Chen Y, Yang J, Cheng Y, Xu L, Ji F, Zhou J, Zhang X, Li Z, Chen C, Ju S, Zhang J, Wang L. Modification of PLAC8 by UFM1 affects tumorous proliferation and immune response by impacting PD-L1 levels in triple-negative breast cancer. J Immunother Cancer 2022; 10:e005668. [PMID: 36543379 PMCID: PMC9772693 DOI: 10.1136/jitc-2022-005668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer is characterized by a poor prognosis and lack of targeted treatments, and thus, new targeting markers and therapeutic strategies are urgently needed. We previously indicated that PLAC8 promotes tumorigenesis and exerts multidrug resistance in breast cancer. Therefore, we aimed to characterize the PLAC8-regulated network in triple-negative breast cancer. METHODS We measured the levels of PLAC8 in breast cancer cell lines and found that PLAC8 is post-translationally modified by ubiquitin-fold modifier 1 (UFM1). Then, we revealed a new regulatory system of PD-L1 by PLAC8 in triple-negative breast cancer. We also tested the molecular functions of PLAC8 in triple-negative breast cancer cell lines and measured the expression of PLAC8 and PD-L1 in breast cancer tissues. RESULTS PLAC8 was generally highly expressed in triple-negative breast cancer and could be modified by UFM1, which maintains PLAC8 protein stability. Moreover, PLAC8 could promote cancer cell proliferation and affect the immune response by regulating the level of PD-L1 ubiquitination. Additionally, among patients with breast cancer, the expression of PLAC8 was higher in triple-negative breast cancer than in non-triple-negative breast cancer and positively correlated with the level of PD-L1. CONCLUSIONS Our current study discoveries a new PLAC8-regulated network in triple-negative breast cancer and provides corresponding guidance for the clinical diagnosis and immunotherapy of triple-negative breast cancer.
Collapse
Affiliation(s)
- Misha Mao
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Yongxia Chen
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Jingjing Yang
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Yifan Cheng
- Department of Gastrointestinal Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Ling Xu
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Feiyang Ji
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Jichun Zhou
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Xun Zhang
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Zhaoqing Li
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Cong Chen
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Siwei Ju
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Jiahang Zhang
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| | - Linbo Wang
- Department of Surgical Oncology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Zhejiang, Hangzhou, China
- Zhejiang university, Zhejiang, Hangzhou, China
| |
Collapse
|