1
|
Han F, Qi G, Li R, Peng J, Yan S, Yuan C, Kong B, Ma H. USP28 promotes PARP inhibitor resistance by enhancing SOX9-mediated DNA damage repair in ovarian cancer. Cell Death Dis 2025; 16:305. [PMID: 40240356 PMCID: PMC12003857 DOI: 10.1038/s41419-025-07647-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
PARP inhibitor (PARPi) resistance presents a significant challenge in ovarian cancer treatment, necessitating the development of effective therapeutic strategies to overcome this resistance and improve patient outcomes. Our study demonstrated that elevated expression of SRY-box 9 (SOX9) contributes to olaparib resistance in ovarian cancer. Mechanistically, the deubiquitinating enzyme USP28 was identified as a novel interacting partner of SOX9. USP28 inhibited the ubiquitination and subsequent degradation of SOX9, which is mediated by the E3 ubiquitin ligase FBXW7 during olaparib treatment. ChIP-Seq analysis revealed that SOX9 binds to the promoters of key DNA damage repair (DDR) genes (SMARCA4, UIMC1, and SLX4), thereby regulating DDR processes in ovarian cancer. Additionally, USP28 promoted olaparib resistance by stabilizing SOX9 protein and enhancing DNA damage repair. Furthermore, the USP28 specific inhibitor AZ1 reduced SOX9 protein stability and increased the sensitivity of ovarian cancer cells to olaparib. In conclusion, targeted inhibition of USP28 promoted ubiquitination-mediated degradation of SOX9, thereby impairing DNA damage repair capabilities and sensitizing ovarian cancer cells to PARPi. These findings elucidate the underlying mechanisms of PARPi resistance in ovarian cancer and suggest the potential efficacy of combining USP28 inhibitors with PARPi to overcome this resistance.
Collapse
Affiliation(s)
- Fang Han
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Gonghua Qi
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Rongrong Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Jiali Peng
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Shi Yan
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Cunzhong Yuan
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hanlin Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
2
|
Jin SK, Baek KH. Unraveling the role of deubiquitinating enzymes on cisplatin resistance in several cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189297. [PMID: 40058507 DOI: 10.1016/j.bbcan.2025.189297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
The use of platinum-based drugs in cancer treatment is one of the most common methods in chemotherapy. Especially, cisplatin induces cell death by interrupting DNA synthesis by binding to the DNA bases, thereby leading to the apoptosis via multiple pathways. However, the major hurdle in chemotherapy is drug resistance. To overcome drug resistance, the ubiquitin-proteasome system (UPS) has emerged as a potential therapeutic target. The UPS is a pivotal signaling pathway that regulates the majority of cellular proteins by attaching ubiquitin to substrates, leading to proteasomal degradation. Conversely, deubiquitinating enzymes (DUBs) remove tagged ubiquitin from the substrate and inhibit degradation, thereby maintaining proteostasis. Recently, studies have been conducted to identify the substrates of DUBs and investigated the cellular mechanisms, and now the development of therapeutics using DUB inhibitors is in clinical trials. However, the mechanism of the DUB response to cisplatin remains still unclear. In this review, we summarize the research reported on the function of DUBs responding to cisplatin.
Collapse
Affiliation(s)
- Sun-Kyu Jin
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea.
| |
Collapse
|
3
|
Zeng L, Li YQ, He SW, Xu H, Zhang R, Chen K, Qin LJ, Zhu XH, Li YL, Li L, Liu N, Wang HY. The deubiquitinase USP44 enhances cisplatin chemosensitivity through stabilizing STUB1 to promote LRPPRC degradation in neuroblastoma. Neuro Oncol 2025; 27:492-507. [PMID: 39215663 PMCID: PMC11812030 DOI: 10.1093/neuonc/noae175] [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: 01/05/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Dysregulated deubiquitinating enzymes (DUBs) execute as intrinsic oncogenes or tumor suppressors and are involved in chemoresistance in cancers. However, the functions and exact molecular mechanisms remain largely unclear in neuroblastoma. METHODS Here, an R2 screening strategy based on the standard deviation values was used to identify the most important DUB, USP44, in neuroblastoma with stage 4. We validated the role of USP44 regulation upon cisplatin treatment in vitro and in vivo experiments, revealing the molecular mechanisms associated with USP44 regulation and cisplatin sensitivity in neuroblastoma. RESULTS We found that low USP44 expression was associated with an inferior prognosis in neuroblastoma patients. Overexpression of USP44 enhanced neuroblastoma cell sensitivity to cisplatin in vitro and in vivo. Mechanistically, USP44 recruited and stabilized the E3 ubiquitin ligase STUB1 by removing its K48-linked polyubiquitin chains at Lys30, and STUB1 further reinforced the K48-linked polyubiquitination of LRPPRC at Lys453 and promoted its protein degradation, thus enhancing the accumulation of mitochondrial reactive oxygen species (mROS), in turn facilitating neuroblastoma cell apoptosis and cisplatin sensitivity. Additionally, overexpression of LRPPRC reversed the promoting effect of USP44 on cell apoptosis in cisplatin-treated neuroblastoma cells. CONCLUSIONS Our findings demonstrate that the USP44-STUB1-LRPPRC axis plays a pivotal role in neuroblastoma chemoresistance and provides potential targets for neuroblastoma therapy and prognostication.
Collapse
Affiliation(s)
- Liang Zeng
- Department of Pathology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children’s Medical Center for South Central Region, Guangzhou, P. R. China
| | - Ying-Qing Li
- Department of Experimental Research, State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Shi-Wei He
- Department of Experimental Research, State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Hui Xu
- Department of Pathology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children’s Medical Center for South Central Region, Guangzhou, P. R. China
| | - Ruizhong Zhang
- Guangdong Provincial Key Laboratory of Research in Structure Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P. R. China
| | - Kai Chen
- Department of Pathology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children’s Medical Center for South Central Region, Guangzhou, P. R. China
| | - Liang-Jun Qin
- Department of Pathology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children’s Medical Center for South Central Region, Guangzhou, P. R. China
| | - Xun-Hua Zhu
- Department of Experimental Research, State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yi-Lin Li
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, P. R. China
| | - Le Li
- Department of Thoracic Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children’s Medical Center for South Central Region, Guangzhou, P. R. China
| | - Na Liu
- Department of Experimental Research, State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Hai-Yun Wang
- Guangzhou Institute of Paediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, National Children’s Medical Center for South Central Region, Guangzhou, P. R. China
- Department of Pathology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children’s Medical Center for South Central Region, Guangzhou, P. R. China
| |
Collapse
|
4
|
Yu D, Zhong Q, Wang Y, Yin C, Bai M, Zhu J, Chen J, Li H, Hong W. Lactylation: The metabolic accomplice shaping cancer's response to radiotherapy and immunotherapy. Ageing Res Rev 2025; 104:102670. [PMID: 39864560 DOI: 10.1016/j.arr.2025.102670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Protein lactylation, an emerging post-translational modification, is providing new insights into tumor biology and challenging our current understanding of cancer mechanisms. Our review illuminates the intricate roles of lactylation in carcinogenesis, tumor progression, and therapeutic responses, positioning it as a critical linchpin connecting metabolic reprogramming, epigenetic modulation, and treatment outcomes. We provide an in-depth analysis of lactylation's molecular mechanisms and its far-reaching impact on cell cycle regulation, immune evasion strategies, and therapeutic resistance within the complex tumor microenvironment. Notably, this review dissects the paradoxical nature of lactylation in cancer immunotherapy and radiotherapy. While heightened lactylation can foster immune suppression and radioresistance, strategically targeting lactylation cascades opens innovative avenues for amplifying the efficacy of current treatment paradigms. We critically evaluate lactylation's potential as a robust diagnostic and prognostic biomarker and explore frontier therapeutic approaches targeting lactylation. The synergistic integration of multi-omics data and artificial intelligence in lactylation research is catalyzing significant strides towards personalized cancer management. This review not only consolidates current knowledge but also charts a course for future investigations. Key research imperatives include deciphering tumor-specific lactylation signatures, optimizing synergistic strategies combining lactylation modulation with immune checkpoint inhibitors and radiotherapy, and comprehensively assessing the long-term physiological implications of lactylation intervention. As our understanding of lactylation's pivotal role in tumor biology continues to evolve, this burgeoning field promises to usher in transformative advancements in cancer diagnosis, treatment modalitie.
Collapse
Affiliation(s)
- Danqing Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Qingping Zhong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yanlin Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Chang Yin
- Nursing Department, Shanghai Sixth People's Hospital, Shanghai 200233, China
| | - Minghua Bai
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ji Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jinggang Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Huaming Li
- Department of Gastroenterology, Hangzhou Third Peoples Hospital, Hangzhou 310000, China.
| | - Weifeng Hong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
5
|
Hira A, Zhang J, Kadakia MP. TIP60 enhances cisplatin resistance via regulating ΔNp63α acetylation in SCC. Cell Death Dis 2024; 15:877. [PMID: 39627186 PMCID: PMC11615348 DOI: 10.1038/s41419-024-07265-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/12/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
Non-melanoma skin cancer, including basal and squamous cell carcinoma, is the most common form of cancer worldwide, with approximately 5.4 million new cases diagnosed each year in the United States. While the chemotherapeutic drug cisplatin is often used to treat squamous cell carcinoma (SCC) patients, low response rates and disease recurrence are common. In this study, we show that TIP60 and ΔNp63α levels correlate with cisplatin resistance in SCC cell lines, suggesting that TIP60 contributes to the failure of platinum-based drugs in SCC by regulating the stability and transcriptional activity of ΔNp63α. Depletion of endogenous TIP60 or pharmacological inhibition of TIP60 led to a decrease in ΔNp63α protein and acetylation levels in multiple SCC cell lines. We showed that TIP60 upregulates ΔNp63α protein levels in cisplatin-resistant SCC cell lines by protecting it from cisplatin-mediated degradation and increasing its protein stability. Stable expression of TIP60 or ΔNp63α individually promoted resistance to cisplatin and reduced cell death, while loss of either TIP60 or ΔNp63α induced G2/M arrest, increased cell death, and sensitized cells to cisplatin. Moreover, pharmacological inhibition of TIP60 reduced acetylation of ΔNp63α and sensitized resistant cells to cisplatin. Taken together, our study indicates that TIP60-mediated stabilization of ΔNp63α increases cisplatin resistance and provides critical insights into the mechanisms by which ΔNp63α confers cisplatin resistance by promoting cell proliferation and inhibiting apoptosis. Furthermore, our data suggests that inhibition of TIP60 may be therapeutically advantageous in overcoming cisplatin resistance in SCC and other epithelial cancers.
Collapse
Affiliation(s)
- Akshay Hira
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Jin Zhang
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Madhavi P Kadakia
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA.
| |
Collapse
|
6
|
Reissland M, Hartmann O, Tauch S, Bugter JM, Prieto-Garcia C, Schulte C, Loebbert S, Solvie D, Bitman-Lotan E, Narain A, Jacomin AC, Schuelein-Voelk C, Fuss CT, Pahor N, Ade C, Buck V, Potente M, Li V, Beliu G, Wiegering A, Grossmann T, Eilers M, Wolf E, Maric H, Rosenfeldt M, Maurice MM, Dikic I, Gallant P, Orian A, Diefenbacher ME. USP10 drives cancer stemness and enables super-competitor signalling in colorectal cancer. Oncogene 2024; 43:3645-3659. [PMID: 39443725 PMCID: PMC11611742 DOI: 10.1038/s41388-024-03141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 07/30/2024] [Accepted: 08/21/2024] [Indexed: 10/25/2024]
Abstract
The contribution of deubiquitylating enzymes (DUBs) to β-Catenin stabilization in intestinal stem cells and colorectal cancer (CRC) is poorly understood. Here, and by using an unbiassed screen, we discovered that the DUB USP10 stabilizes β-Catenin specifically in APC-truncated CRC in vitro and in vivo. Mechanistic studies, including in vitro binding together with computational modelling, revealed that USP10 binding to β-Catenin is mediated via the unstructured N-terminus of USP10 and is outcompeted by intact APC, favouring β-catenin degradation. However, in APC-truncated cancer cells USP10 binds to β-catenin, increasing its stability which is critical for maintaining an undifferentiated tumour identity. Elimination of USP10 reduces the expression of WNT and stem cell signatures and induces the expression of differentiation genes. Remarkably, silencing of USP10 in murine and patient-derived CRC organoids established that it is essential for NOTUM signalling and the APC super competitor-phenotype, reducing tumorigenic properties of APC-truncated CRC. These findings are clinically relevant as patient-derived organoids are highly dependent on USP10, and abundance of USP10 correlates with poorer prognosis of CRC patients. Our findings reveal, therefore, a role for USP10 in CRC cell identity, stemness, and tumorigenic growth by stabilising β-Catenin, leading to aberrant WNT signalling and degradation resistant tumours. Thus, USP10 emerges as a unique therapeutic target in APC truncated CRC.
Collapse
Affiliation(s)
- Michaela Reissland
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Oliver Hartmann
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
| | - Saskia Tauch
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen M Bugter
- Oncode Institute and Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Cristian Prieto-Garcia
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Molecular Signalling Group, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Clemens Schulte
- Rudolf-Virchow-Center for Integrative and Translational Imaging, University of Wuerzburg, Wuerzburg, Germany
| | - Sinah Loebbert
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Daniel Solvie
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Ashwin Narain
- Cancer Systems Biology Group, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Anne-Claire Jacomin
- Molecular Signalling Group, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Carmina T Fuss
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Nikolett Pahor
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
| | - Carsten Ade
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Viktoria Buck
- Institute for Pathology, University of Würzburg, Wuerzburg, Germany
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin Germany and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Vivian Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Gerti Beliu
- Rudolf-Virchow-Center for Integrative and Translational Imaging, University of Wuerzburg, Wuerzburg, Germany
| | - Armin Wiegering
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
- Department of General, Visceral, Transplantation, Vascular and Paediatric Surgery, University Hospital, Julius-Maximilians-University of Wuerzburg, Wuerzburg, Germany
| | - Tom Grossmann
- Amsterdam Institute of Molecular and Life Sciences, Amsterdam, Netherlands
| | - Martin Eilers
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Elmar Wolf
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Cancer Systems Biology Group, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Hans Maric
- Rudolf-Virchow-Center for Integrative and Translational Imaging, University of Wuerzburg, Wuerzburg, Germany
| | - Mathias Rosenfeldt
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Institute for Pathology, University of Würzburg, Wuerzburg, Germany
| | - Madelon M Maurice
- Oncode Institute and Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ivan Dikic
- Molecular Signalling Group, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Peter Gallant
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Amir Orian
- Faculty of Medicine, TICC, Technion Haifa, Haifa, Israel
| | - Markus E Diefenbacher
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany.
- Institute of Lung Health and Immunity, Helmholtz Center, Munich, Germany.
- German Center for Lung Research, DZL, Germany.
- Ludwig Maximilian University Munich, Munich, Germany.
| |
Collapse
|
7
|
Prieto-Garcia C, Matkovic V, Mosler T, Li C, Liang J, Oo JA, Haidle F, Mačinković I, Cabrera-Orefice A, Berkane R, Giuliani G, Xu F, Jacomin AC, Tomaskovic I, Basoglu M, Hoffmann ME, Rathore R, Cetin R, Boutguetait D, Bozkurt S, Hernández Cañás MC, Keller M, Busam J, Shah VJ, Wittig I, Kaulich M, Beli P, Galej WP, Ebersberger I, Wang L, Münch C, Stolz A, Brandes RP, Tse WKF, Eimer S, Stainier DYR, Legewie S, Zarnack K, Müller-McNicoll M, Dikic I. Pathogenic proteotoxicity of cryptic splicing is alleviated by ubiquitination and ER-phagy. Science 2024; 386:768-776. [PMID: 39541449 DOI: 10.1126/science.adi5295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/22/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
RNA splicing enables the functional adaptation of cells to changing contexts. Impaired splicing has been associated with diseases, including retinitis pigmentosa, but the underlying molecular mechanisms and cellular responses remain poorly understood. In this work, we report that deficiency of ubiquitin-specific protease 39 (USP39) in human cell lines, zebrafish larvae, and mice led to impaired spliceosome assembly and a cytotoxic splicing profile characterized by the use of cryptic 5' splice sites. Disruptive cryptic variants evaded messenger RNA (mRNA) surveillance pathways and were translated into misfolded proteins, which caused proteotoxic aggregates, endoplasmic reticulum (ER) stress, and, ultimately, cell death. The detrimental consequence of splicing-induced proteotoxicity could be mitigated by up-regulating the ubiquitin-proteasome system and selective autophagy. Our findings provide insight into the molecular pathogenesis of spliceosome-associated diseases.
Collapse
Affiliation(s)
- Cristian Prieto-Garcia
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Vigor Matkovic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Thorsten Mosler
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Congxin Li
- Department of Systems Biology and Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - Jie Liang
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - James A Oo
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Goethe University Frankfurt, Frankfurt, Germany
| | - Felix Haidle
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Igor Mačinković
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Alfredo Cabrera-Orefice
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
- Center for Functional Proteomics, Goethe University Frankfurt, Frankfurt, Germany
| | - Rayene Berkane
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Giulio Giuliani
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Fenfen Xu
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Anne-Claire Jacomin
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Ines Tomaskovic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Marion Basoglu
- Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
| | - Marina E Hoffmann
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Rajeshwari Rathore
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Ronay Cetin
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Doha Boutguetait
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Süleyman Bozkurt
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Mario Keller
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Jonas Busam
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Varun Jayeshkumar Shah
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Ilka Wittig
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany
- Center for Functional Proteomics, Goethe University Frankfurt, Frankfurt, Germany
| | - Manuel Kaulich
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Petra Beli
- Institute of Molecular Biology (IMB), Mainz, Germany
- Institute of Developmental Biology and Neurobiology (IDN), Johannes Gutenberg-University, Mainz, Germany
| | | | - Ingo Ebersberger
- Applied Bioinformatics Group, Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
- Senckenberg Biodiversity and Climate Research Centre (S-BIK-F), Frankfurt, Germany
- LOEWE Centre for Translational Biodiversity Genomics (TBG), Frankfurt, Germany
| | - Likun Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Christian Münch
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Center for Functional Proteomics, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Alexandra Stolz
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Goethe University Frankfurt, Frankfurt, Germany
| | - William Ka Fai Tse
- Laboratory of Developmental Disorders and Toxicology, Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Stefan Eimer
- Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Stefan Legewie
- Department of Systems Biology and Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - Kathi Zarnack
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Michaela Müller-McNicoll
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
- Max-Planck Institute for Biophysics, Frankfurt, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Max-Planck Institute for Biophysics, Frankfurt, Germany
| |
Collapse
|
8
|
Jin C, Einig E, Xu W, Kollampally RB, Schlosser A, Flentje M, Popov N. The dimeric deubiquitinase USP28 integrates 53BP1 and MYC functions to limit DNA damage. Nucleic Acids Res 2024; 52:3011-3030. [PMID: 38227944 PMCID: PMC11024517 DOI: 10.1093/nar/gkae004] [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: 04/14/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024] Open
Abstract
DNA replication is a major source of endogenous DNA damage in tumor cells and a key target of cellular response to genotoxic stress. DNA replication can be deregulated by oncoproteins, such as transcription factor MYC, aberrantly activated in many human cancers. MYC is stringently regulated by the ubiquitin system - for example, ubiquitination controls recruitment of the elongation factor PAF1c, instrumental in MYC activity. Curiously, a key MYC-targeting deubiquitinase USP28 also controls cellular response to DNA damage via the mediator protein 53BP1. USP28 forms stable dimers, but the biological role of USP28 dimerization is unknown. We show here that dimerization limits USP28 activity and restricts recruitment of PAF1c by MYC. Expression of monomeric USP28 stabilizes MYC and promotes PAF1c recruitment, leading to ectopic DNA synthesis and replication-associated DNA damage. USP28 dimerization is stimulated by 53BP1, which selectively binds USP28 dimers. Genotoxic stress diminishes 53BP1-USP28 interaction, promotes disassembly of USP28 dimers and stimulates PAF1c recruitment by MYC. This triggers firing of DNA replication origins during early response to genotoxins and exacerbates DNA damage. We propose that dimerization of USP28 prevents ectopic DNA replication at transcriptionally active chromatin to maintain genome stability.
Collapse
Affiliation(s)
- Chao Jin
- Department of Medical Oncology and Pulmonology, University Hospital Tübingen, Otfried-Müller-Str 14, 72076 Tübingen, Germany
- DFG Cluster of Excellence 2180 ‘Image-guided and Functionally Instructed Tumor Therapies’ (iFIT), University of Tübingen, Tübingen, Germany
| | - Elias Einig
- Department of Medical Oncology and Pulmonology, University Hospital Tübingen, Otfried-Müller-Str 14, 72076 Tübingen, Germany
- DFG Cluster of Excellence 2180 ‘Image-guided and Functionally Instructed Tumor Therapies’ (iFIT), University of Tübingen, Tübingen, Germany
| | - Wenshan Xu
- Department of Radiation Oncology, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Ravi Babu Kollampally
- Department of Medical Oncology and Pulmonology, University Hospital Tübingen, Otfried-Müller-Str 14, 72076 Tübingen, Germany
- DFG Cluster of Excellence 2180 ‘Image-guided and Functionally Instructed Tumor Therapies’ (iFIT), University of Tübingen, Tübingen, Germany
| | - Andreas Schlosser
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str 2, 97080 Würzburg, Germany
| | - Michael Flentje
- Department of Radiation Oncology, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Nikita Popov
- Department of Medical Oncology and Pulmonology, University Hospital Tübingen, Otfried-Müller-Str 14, 72076 Tübingen, Germany
- DFG Cluster of Excellence 2180 ‘Image-guided and Functionally Instructed Tumor Therapies’ (iFIT), University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Karapurkar JK, Colaco JC, Suresh B, Tyagi A, Woo SH, Jo WJ, Ko N, Singh V, Hong SH, Oh SJ, Kim KS, Ramakrishna S. USP28 promotes tumorigenesis and cisplatin resistance by deubiquitinating MAST1 protein in cancer cells. Cell Mol Life Sci 2024; 81:145. [PMID: 38498222 PMCID: PMC10948558 DOI: 10.1007/s00018-024-05187-2] [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: 08/01/2023] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
Cisplatin is a chemotherapy drug that causes a plethora of DNA lesions and inhibits DNA transcription and replication, resulting in the induction of apoptosis in cancer cells. However, over time, patients develop resistance to cisplatin due to repeated treatment and thus the treatment efficacy is limited. Therefore, identifying an alternative therapeutic strategy combining cisplatin treatment along with targeting factors that drive cisplatin resistance is needed. CRISPR/Cas9 system-based genome-wide screening for the deubiquitinating enzyme (DUB) subfamily identified USP28 as a potential DUB that governs cisplatin resistance. USP28 regulates the protein level of microtubule-associated serine/threonine kinase 1 (MAST1), a common kinase whose expression is elevated in several cisplatin-resistant cancer cells. The expression level and protein turnover of MAST1 is a major factor driving cisplatin resistance in many cancer types. Here we report that the USP28 interacts and extends the half-life of MAST1 protein by its deubiquitinating activity. The expression pattern of USP28 and MAST1 showed a positive correlation across a panel of tested cancer cell lines and human clinical tissues. Additionally, CRISPR/Cas9-mediated gene knockout of USP28 in A549 and NCI-H1299 cells blocked MAST1-driven cisplatin resistance, resulting in suppressed cell proliferation, colony formation ability, migration and invasion in vitro. Finally, loss of USP28 destabilized MAST1 protein and attenuated tumor growth by sensitizing cells to cisplatin treatment in mouse xenograft model. We envision that targeting the USP28-MAST1 axis along with cisplatin treatment might be an alternative therapeutic strategy to overcome cisplatin resistance in cancer patients.
Collapse
Affiliation(s)
| | - Jencia Carminha Colaco
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Bharathi Suresh
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Apoorvi Tyagi
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Sang Hyeon Woo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Won-Jun Jo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Nare Ko
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, 05505, Korea
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea.
- College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea.
- College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| |
Collapse
|
10
|
Xu Z, Wang H, Meng Q, Ding Y, Zhu M, Zhou H, Zhang N, Shi L. Otilonium Bromide acts as a selective USP28 inhibitor and exhibits cytotoxic activity against multiple human cancer cell lines. Biochem Pharmacol 2023; 215:115746. [PMID: 37579857 DOI: 10.1016/j.bcp.2023.115746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
USP28 contributes to tumorigenesis through modulating the lifespan of oncogenic factors such as c-Myc and ΔNp63, and it has been identified as a potential target for anti-cancer drug development. Currently, although quite a number of USP28 inhibitors have been developed, they all are still in preclinical research stage. Besides, none of them exhibits satisfying inhibition selectivity against USP28 over its closest homologue USP25. Here in this manuscript, a high-throughput screening aiming to discover USP28 inhibitors with novel scaffold and enhanced inhibition selectivity were conducted. After the primary screening and the second round of validation, Otilonium Bromide, an approved drug for treating irritable bowel syndrome, was identified to inhibit USP28's activity with the IC50 value at 6.90 ± 0.90 μM. Besides, the drug exhibits a 3-4 folds inhibition selectivity against USP28 over USP25. According to the enzymatic kinetics analysis data and the hydrogen-deuterium exchange mass spectrometry results, Otilonium Bromide could bind to the allosteric pocket of USP28 and inhibit its activity in a reversible and non-competitive mode. The following performed cell-based assays revealed that the drug could cause cytotoxicity against human colorectal cancer cells and lung squamous carcinoma cells potentially through down-regulating USP28's oncogenic substrates c-Myc and/or ΔNp63. Meanwhile, since Otilonium Bromide has been found to preferentially distribute to gastrointestinal tissues, we then evaluated its potential in the combination treatment of colorectal cancer cells with Regorafenib, which is an approved drug for colorectal cancer therapy. As expected, Otilonium Bromide could significantly enhance the sensitivity of colorectal cancer cells to Regorafenib.
Collapse
Affiliation(s)
- Zhuo Xu
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Hui Wang
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Qian Meng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Yiluan Ding
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Mengying Zhu
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Hu Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Naixia Zhang
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China.
| | - Li Shi
- State Key Laboratory of Chemical Biology, Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.
| |
Collapse
|
11
|
Zhou W, Chen J, Wang J. Comprehensive prognostic and immunological analysis of Ubiquitin Specific Peptidase 28 in pan-cancers and identification of its role in hepatocellular carcinoma cell lines. Aging (Albany NY) 2023; 15:6545-6576. [PMID: 37450415 PMCID: PMC10373984 DOI: 10.18632/aging.204869] [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: 04/24/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Ubiquitin Specific Peptidase 28 (USP28), as a member of the DUBs family, has been reported to regulate the occurrence and development of some tumors, but its oncogenic role in tumor immunity is still unknown. METHODS The comprehensive view of USP28 expression in tumor and normal samples was obtained from public databases, including The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Cancer Cell Line Encyclopedia (CCLE). We analyzed the genomic alterations of USP28 in various cancers using the cBioPortal dataset. Besides, gene set enrichment analysis was used to analyze the associated cancer hallmarks with USP28 expression, and TIMER2.0 was taken to investigate the immune cell infiltrations related to the USP28 level. RESULTS USP28 is highly expressed in most tumors and has prognostic value across various cancer types. Moreover, a significant correlation exists between USP28 and immune regulators, clinical staging, checkpoint inhibitor response, MSI, TMB, CNV, MMR defects, and DNA methylation. Additionally, USP28 expression is strongly associated with the infiltration levels of neutrophils and NK cells in most tumor types. One of the most significant findings of our study was that USP28 could serve as a significant predictor of anti-CTLA4 therapy response in melanoma patients. Additionally, our molecular biology experiments validated that the knockdown of USP28 substantially reduced the proliferative and invasive abilities of the HCC cell lines. CONCLUSIONS Our study suggests that USP28 could potentially serve as a biomarker for cancer immunologic infiltration and poor prognosis, with potential applications in developing novel cancer treatment strategies.
Collapse
Affiliation(s)
- Wuhan Zhou
- Department of Hepatobiliary Surgery, The First Hospital of Putian City, Putian 351100, Fujian, China
| | - Jiafei Chen
- Department of Hepatobiliary Surgery, The First Hospital of Putian City, Putian 351100, Fujian, China
| | - Jingui Wang
- Department of Hepatobiliary Surgery, The First Hospital of Putian City, Putian 351100, Fujian, China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou 350108, Fujian, China
| |
Collapse
|
12
|
Maier CR, Hartmann O, Prieto-Garcia C, Al-Shami KM, Schlicker L, Vogel FCE, Haid S, Klann K, Buck V, Münch C, Schmitz W, Einig E, Krenz B, Calzado MA, Eilers M, Popov N, Rosenfeldt MT, Diefenbacher ME, Schulze A. USP28 controls SREBP2 and the mevalonate pathway to drive tumour growth in squamous cancer. Cell Death Differ 2023:10.1038/s41418-023-01173-6. [PMID: 37202505 DOI: 10.1038/s41418-023-01173-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/16/2023] [Accepted: 04/25/2023] [Indexed: 05/20/2023] Open
Abstract
SREBP2 is a master regulator of the mevalonate pathway (MVP), a biosynthetic process that drives the synthesis of dolichol, heme A, ubiquinone and cholesterol and also provides substrates for protein prenylation. Here, we identify SREBP2 as a novel substrate for USP28, a deubiquitinating enzyme that is frequently upregulated in squamous cancers. Our results show that silencing of USP28 reduces expression of MVP enzymes and lowers metabolic flux into this pathway. We also show that USP28 binds to mature SREBP2, leading to its deubiquitination and stabilisation. USP28 depletion rendered cancer cells highly sensitive to MVP inhibition by statins, which was rescued by the addition of geranyl-geranyl pyrophosphate. Analysis of human tissue microarrays revealed elevated expression of USP28, SREBP2 and MVP enzymes in lung squamous cell carcinoma (LSCC) compared to lung adenocarcinoma (LADC). Moreover, CRISPR/Cas-mediated deletion of SREBP2 selectively attenuated tumour growth in a KRas/p53/LKB1 mutant mouse model of lung cancer. Finally, we demonstrate that statins synergise with a dual USP28/25 inhibitor to reduce viability of SCC cells. Our findings suggest that combinatorial targeting of MVP and USP28 could be a therapeutic strategy for the treatment of squamous cell carcinomas.
Collapse
Affiliation(s)
- Carina R Maier
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- Protein Stability and Cancer Group, Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany
| | - Oliver Hartmann
- Protein Stability and Cancer Group, Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany
| | - Cristian Prieto-Garcia
- Protein Stability and Cancer Group, Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany
- Institute of Biochemistry II, Goethe University Frankfurt, Theodor-Stern-Kai 7, Haus 75, 60590, Frankfurt am Main, Germany
| | - Kamal M Al-Shami
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Lisa Schlicker
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Felix C E Vogel
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Silke Haid
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Kevin Klann
- Institute of Biochemistry II, Goethe University Frankfurt, Theodor-Stern-Kai 7, Haus 75, 60590, Frankfurt am Main, Germany
| | - Viktoria Buck
- Institute of Pathology, Julius Maximilians University and Comprehensive Cancer Center (CCC) Mainfranken, Josef-Schneider-Strasse 2, 97080, Würzburg, Germany
| | - Christian Münch
- Institute of Biochemistry II, Goethe University Frankfurt, Theodor-Stern-Kai 7, Haus 75, 60590, Frankfurt am Main, Germany
| | - Werner Schmitz
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Elias Einig
- Internal Medicine VIII-Clinical Tumor Biology, University of Tübingen, Otfried-Müller-Straße 14, 72076, Tübingen, Germany
| | - Bastian Krenz
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Marco A Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Martin Eilers
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Nikita Popov
- Internal Medicine VIII-Clinical Tumor Biology, University of Tübingen, Otfried-Müller-Straße 14, 72076, Tübingen, Germany
| | - Mathias T Rosenfeldt
- Institute of Pathology, Julius Maximilians University and Comprehensive Cancer Center (CCC) Mainfranken, Josef-Schneider-Strasse 2, 97080, Würzburg, Germany
| | - Markus E Diefenbacher
- Protein Stability and Cancer Group, Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany.
| | - Almut Schulze
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany.
| |
Collapse
|
13
|
Zhou D, Xu Z, Huang Y, Wang H, Zhu X, Zhang W, Song W, Gao T, Liu T, Wang M, Shi L, Zhang N, Xiong B. Structure-based discovery of potent USP28 inhibitors derived from Vismodegib. Eur J Med Chem 2023; 254:115369. [PMID: 37075624 DOI: 10.1016/j.ejmech.2023.115369] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 04/21/2023]
Abstract
Ubiquitin-specific proteases (USPs) 28 is overexpressed in multiple types of cancers. The development of potent USP28 inhibitors is still in primitive stage. We previously reported our discovery of Vismodegib as a USP28 inhibitor by screening a commercially available drug library. Herein, we report our efforts to solve the cocrystal structure of Vismodegib bound to USP28 for the first time and subsequent structure-based optimization leading to a series of Vismodegib derivatives as potent USP28 inhibitors. Based on the cocrystal structure, elaborative SARs exploration was carried out to afford much more potent USP28 inhibitors than Vismodegib. The representative compounds 9l, 9o and 9p bearing high potency on USP28 showed high selectivity over USP2, USP7, USP8, USP9x, UCHL3 and UCHL5. The detailed cellular assay suggested that compounds 9l, 9o and 9p could cause cytotoxicity in both human colorectal cancer and lung squamous carcinoma cells and significantly enhance the sensitivity of colorectal cancer cells to Regorafenib. Further immunoblotting analysis indicated that compounds 9l, 9o and 9p could dose-dependently down-regulate the cellular level of c-Myc through ubiquitin-proteasome system and anti-cancer effects could mainly be attributed to their inhibition on USP28 but not involving the Hedgehog-Smoothened pathway. Thus, our work provided a series of novel and potent USP28 inhibitors derived from Vismodegib and may contribute to the development of USP28 inhibitors.
Collapse
Affiliation(s)
- Di Zhou
- Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan District, Hefei, Anhui, 230012, PR China; Yangtze Delta Drug Advnced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China
| | - Zhuo Xu
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, PR China
| | - Yaodong Huang
- Shenyang Pharmaceutical University, 103 Wenhua Rd, Shenyang, Liaoning, 110016, PR China; Yangtze Delta Drug Advnced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China
| | - Hui Wang
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China
| | - Xiaoli Zhu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; Yangtze Delta Drug Advnced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China
| | - Wentao Zhang
- Shenyang Pharmaceutical University, 103 Wenhua Rd, Shenyang, Liaoning, 110016, PR China; Yangtze Delta Drug Advnced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China
| | - Weiwei Song
- Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan District, Hefei, Anhui, 230012, PR China; Yangtze Delta Drug Advnced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China
| | - Tong Gao
- Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan District, Hefei, Anhui, 230012, PR China; Yangtze Delta Drug Advnced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China
| | - Tongchao Liu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China
| | - Meng Wang
- Shanghai Chemvon Biotechnology Company (Limited), Shanghai, 201202, PR China; Yangtze Delta Drug Advnced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China.
| | - Li Shi
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China.
| | - Naixia Zhang
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, PR China.
| | - Bing Xiong
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, PR China; Yangtze Delta Drug Advnced Research Institute, 100 Dongtinghu Road, Nantong, 226133, PR China.
| |
Collapse
|
14
|
Zhao Z, Xu H, Wei Y, Sun L, Song Y. Deubiquitylase PSMD14 inhibits autophagy to promote ovarian cancer progression via stabilization of LRPPRC. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166594. [PMID: 36328147 DOI: 10.1016/j.bbadis.2022.166594] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022]
Abstract
Autophagy is a key cellular process, which exists in many tumors and plays dual roles in tumor promotion and suppression. However, the role and mechanism of aberrant autophagy in ovarian cancer remains unclear. Ubiquitin-proteasome pathway is the most important pathway for specific protein degradation. Deubiquitinases (DUBs) have crucial roles in all the stages of tumorigenesis and progression. Herein, we explore the DUBs which contribute to aberrant autophagy in ovarian cancer. TCGA data analysis shows that the autophagy level is suppressed, and the selective autophagy receptor SQSTM1/p62 is abnormally high expressed in ovarian cancer. We screen and identify that the deubiquitinase PSMD14 negatively regulates autophagy level. Functional studies show that increased PSMD14 expression remarkably enhances ovarian cancer cells malignancy, whereas knockdown of PSMD14 has the opposite effect. Furthermore, in vivo assays show that knockdown of PSMD14 inhibits the growth, lung and abdominal metastasis of ovarian cancer. Mechanistically, PSMD14 directly interacts with LRPPRC and inhibits its ubiquitination, thereby inhibiting autophagy through LRPPRC/Beclin1-Bcl-2/SQSTM1 signaling pathway. Next, we demonstrate that PSMD14 is upregulated in ovarian cancer and high expression of PSMD14 positively correlates with LRPPRC. Taken together, we clarify the role of autophagy in regulating the ovarian cancer phenotype and provide insights into regulatory mechanism of autophagy in ovarian cancer.
Collapse
Affiliation(s)
- Zitong Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Heyang Xu
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yuan Wei
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Sun
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China; Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
15
|
Shen J, Xie M, Xu Y, Qian Q, Qiu T, Shi W, Ren D, Ji J, Huang J. Identification of the deubiquitinase USP28 as a novel molecular therapeutic target of ovarian cancer. Biochem Biophys Res Commun 2023; 638:184-191. [PMID: 36462492 DOI: 10.1016/j.bbrc.2022.11.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/12/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Ubiquitin specific proteinase 28 (USP28) is a member of the deubiquitylating enzymes, which are mainly involved in cell cycle, apoptosis and DNA damage repair. Although USP28 has been found to be upregulated in some tumors, its role in ovarian cancer (OV) remains unclear. Here we show that USP28 was highly expressed in OV samples compared with normal ovarian tissue, and OV patients with higher USP28 levels had a worse prognosis. We found that the abnormal expression of USP28 mRNA in OV was related to the activation of β-catenin signaling pathway, and USP28 was a transcriptional target gene of the β-catenin/YAP1/TBX5 complex. In addition, genetic ablation or pharmacological inhibition of USP28 impaired the proliferation ability of OV cells in vitro and in vivo. In conclusion, our findings show that β-catenin/YAP1/TBX5-mediated aberrant expression of USP28 promotes the malignant phenotype of OV, suggesting that USP28 may be a therapeutic target for OV.
Collapse
Affiliation(s)
- Jing Shen
- Department of Obstetrics and Gynecology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Mengru Xie
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yuxin Xu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Qilan Qian
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Teng Qiu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wen Shi
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Dexu Ren
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Jinling Huang
- Department of Obstetrics & Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
16
|
Xu Y, Yang X, Xiong Q, Han J, Zhu Q. The dual role of p63 in cancer. Front Oncol 2023; 13:1116061. [PMID: 37182132 PMCID: PMC10174455 DOI: 10.3389/fonc.2023.1116061] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
The p53 family is made up of three transcription factors: p53, p63, and p73. These proteins are well-known regulators of cell function and play a crucial role in controlling various processes related to cancer progression, including cell division, proliferation, genomic stability, cell cycle arrest, senescence, and apoptosis. In response to extra- or intracellular stress or oncogenic stimulation, all members of the p53 family are mutated in structure or altered in expression levels to affect the signaling network, coordinating many other pivotal cellular processes. P63 exists as two main isoforms (TAp63 and ΔNp63) that have been contrastingly discovered; the TA and ΔN isoforms exhibit distinguished properties by promoting or inhibiting cancer progression. As such, p63 isoforms comprise a fully mysterious and challenging regulatory pathway. Recent studies have revealed the intricate role of p63 in regulating the DNA damage response (DDR) and its impact on diverse cellular processes. In this review, we will highlight the significance of how p63 isoforms respond to DNA damage and cancer stem cells, as well as the dual role of TAp63 and ΔNp63 in cancer.
Collapse
Affiliation(s)
- Yongfeng Xu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaojuan Yang
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qunli Xiong
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qing Zhu, ; Junhong Han,
| | - Qing Zhu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Qing Zhu, ; Junhong Han,
| |
Collapse
|
17
|
Chen J, Wang X, Ma A, Wang QE, Liu B, Li L, Xu D, Ma Q. Deep transfer learning of cancer drug responses by integrating bulk and single-cell RNA-seq data. Nat Commun 2022; 13:6494. [PMID: 36310235 PMCID: PMC9618578 DOI: 10.1038/s41467-022-34277-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/19/2022] [Indexed: 12/25/2022] Open
Abstract
Drug screening data from massive bulk gene expression databases can be analyzed to determine the optimal clinical application of cancer drugs. The growing amount of single-cell RNA sequencing (scRNA-seq) data also provides insights into improving therapeutic effectiveness by helping to study the heterogeneity of drug responses for cancer cell subpopulations. Developing computational approaches to predict and interpret cancer drug response in single-cell data collected from clinical samples can be very useful. We propose scDEAL, a deep transfer learning framework for cancer drug response prediction at the single-cell level by integrating large-scale bulk cell-line data. The highlight in scDEAL involves harmonizing drug-related bulk RNA-seq data with scRNA-seq data and transferring the model trained on bulk RNA-seq data to predict drug responses in scRNA-seq. Another feature of scDEAL is the integrated gradient feature interpretation to infer the signature genes of drug resistance mechanisms. We benchmark scDEAL on six scRNA-seq datasets and demonstrate its model interpretability via three case studies focusing on drug response label prediction, gene signature identification, and pseudotime analysis. We believe that scDEAL could help study cell reprogramming, drug selection, and repurposing for improving therapeutic efficacy.
Collapse
Affiliation(s)
- Junyi Chen
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Xiaoying Wang
- Department of Mathematics, Shandong University, Shandong, 250100, China
| | - Anjun Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.
| | - Qi-En Wang
- Department of Radiation Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Bingqiang Liu
- Department of Mathematics, Shandong University, Shandong, 250100, China
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
18
|
Signaling pathways and targeted therapies in lung squamous cell carcinoma: mechanisms and clinical trials. Signal Transduct Target Ther 2022; 7:353. [PMID: 36198685 PMCID: PMC9535022 DOI: 10.1038/s41392-022-01200-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/03/2022] [Accepted: 09/18/2022] [Indexed: 11/08/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death across the world. Unlike lung adenocarcinoma, patients with lung squamous cell carcinoma (LSCC) have not benefitted from targeted therapies. Although immunotherapy has significantly improved cancer patients' outcomes, the relatively low response rate and severe adverse events hinder the clinical application of this promising treatment in LSCC. Therefore, it is of vital importance to have a better understanding of the mechanisms underlying the pathogenesis of LSCC as well as the inner connection among different signaling pathways, which will surely provide opportunities for more effective therapeutic interventions for LSCC. In this review, new insights were given about classical signaling pathways which have been proved in other cancer types but not in LSCC, including PI3K signaling pathway, VEGF/VEGFR signaling, and CDK4/6 pathway. Other signaling pathways which may have therapeutic potentials in LSCC were also discussed, including the FGFR1 pathway, EGFR pathway, and KEAP1/NRF2 pathway. Next, chromosome 3q, which harbors two key squamous differentiation markers SOX2 and TP63 is discussed as well as its related potential therapeutic targets. We also provided some progress of LSCC in epigenetic therapies and immune checkpoints blockade (ICB) therapies. Subsequently, we outlined some combination strategies of ICB therapies and other targeted therapies. Finally, prospects and challenges were given related to the exploration and application of novel therapeutic strategies for LSCC.
Collapse
|
19
|
Novak R, Ahmad YA, Timaner M, Bitman-Lotan E, Oknin-Vaisman A, Horwitz R, Hartmann O, Reissland M, Buck V, Rosenfeldt M, Nikomarov D, Diefenbacher ME, Shaked Y, Orian A. RNF4~RGMb~BMP6 axis required for osteogenic differentiation and cancer cell survival. Cell Death Dis 2022; 13:820. [PMID: 36153321 PMCID: PMC9509360 DOI: 10.1038/s41419-022-05262-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 01/23/2023]
Abstract
Molecular understanding of osteogenic differentiation (OD) of human bone marrow-derived mesenchymal stem cells (hBMSCs) is important for regenerative medicine and has direct implications for cancer. We report that the RNF4 ubiquitin ligase is essential for OD of hBMSCs, and that RNF4-deficient hBMSCs remain as stalled progenitors. Remarkably, incubation of RNF4-deficient hBMSCs in conditioned media of differentiating hBMSCs restored OD. Transcriptional analysis of RNF4-dependent gene signatures identified two secreted factors that act downstream of RNF4 promoting OD: (1) BMP6 and (2) the BMP6 co-receptor, RGMb (Dragon). Indeed, knockdown of either RGMb or BMP6 in hBMSCs halted OD, while only the combined co-addition of purified RGMb and BMP6 proteins to RNF4-deficient hBMSCs fully restored OD. Moreover, we found that the RNF4-RGMb-BMP6 axis is essential for survival and tumorigenicity of osteosarcoma and therapy-resistant melanoma cells. Importantly, patient-derived sarcomas such as osteosarcoma, Ewing sarcoma, liposarcomas, and leiomyosarcomas exhibit high levels of RNF4 and BMP6, which are associated with reduced patient survival. Overall, we discovered that the RNF4~BMP6~RGMb axis is required for both OD and tumorigenesis.
Collapse
Affiliation(s)
- Rostislav Novak
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel ,Rambam Health Campus Center, Haifa, 3109610 Israel
| | - Yamen Abu Ahmad
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Michael Timaner
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Eliya Bitman-Lotan
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Avital Oknin-Vaisman
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Roi Horwitz
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Oliver Hartmann
- grid.8379.50000 0001 1958 8658Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Michaela Reissland
- grid.8379.50000 0001 1958 8658Protein Stability and Cancer Group, University of Würzburg, Department of Biochemistry and Molecular Biology, Würzburg, Germany
| | - Viktoria Buck
- grid.8379.50000 0001 1958 8658Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Mathias Rosenfeldt
- grid.8379.50000 0001 1958 8658Department of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Markus Elmar Diefenbacher
- grid.8379.50000 0001 1958 8658Protein Stability and Cancer Group, University of Würzburg, Department of Biochemistry and Molecular Biology, Würzburg, Germany
| | - Yuval Shaked
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Amir Orian
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| |
Collapse
|
20
|
Sun X, Cai M, Wu L, Zhen X, Chen Y, Peng J, Han S, Zhang P. USP28 Deubiquitinates TCF7L2 to Govern the Action of Wnt Signaling Pathway in Hepatic Carcinoma. Cancer Sci 2022; 113:3463-3475. [PMID: 35880246 PMCID: PMC9530868 DOI: 10.1111/cas.15509] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022] Open
Abstract
Overexpression of ubiquitin‐specific protease 28 (USP28) is found in hepatic carcinoma. It is unclear whether the deubiquitinase plays a role in hepatocarcinogenesis. Deregulation of the Wnt signaling pathway is frequently associated with liver cancer. Transcription factor 7‐like 2 (TCF7L2) is an important downstream transcription factor of the Wnt/β‐catenin signaling pathway, but the mechanisms by which TCF7L2 itself is regulated have not yet been revealed. Here, we report that USP28 promotes the activity of the Wnt signaling pathway through maintaining the stability of TCF7L2. We further show that FBXW7 is the E3 ubiquitin ligase for TCF7L2. By regulating the levels of TCF7L2, USP28 modulates the Wnt/β‐catenin signaling in liver cancer and USP28 depletion or inhibition by a small molecule inhibitor leads to a halt of growth in liver cancer cells. These results suggest that USP28 could be a potential therapeutic target for liver cancer.
Collapse
Affiliation(s)
- Xiao Sun
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengjiao Cai
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, China
| | - Lingzhi Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinghua Zhen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuetong Chen
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, China
| | - Jin Peng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Suxia Han
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, China
| | - Pumin Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University Medical School, Hangzhou, Zhejiang, China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Ali R, Aouida M, Alhaj Sulaiman A, Madhusudan S, Ramotar D. Can Cisplatin Therapy Be Improved? Pathways That Can Be Targeted. Int J Mol Sci 2022; 23:ijms23137241. [PMID: 35806243 PMCID: PMC9266583 DOI: 10.3390/ijms23137241] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 02/04/2023] Open
Abstract
Cisplatin (cis-diamminedichloroplatinum (II)) is the oldest known chemotherapeutic agent. Since the identification of its anti-tumour activity, it earned a remarkable place as a treatment of choice for several cancer types. It remains effective against testicular, bladder, lung, head and neck, ovarian, and other cancers. Cisplatin treatment triggers different cellular responses. However, it exerts its cytotoxic effects by generating inter-strand and intra-strand crosslinks in DNA. Tumour cells often develop tolerance mechanisms by effectively repairing cisplatin-induced DNA lesions or tolerate the damage by adopting translesion DNA synthesis. Cisplatin-associated nephrotoxicity is also a huge challenge for effective therapy. Several preclinical and clinical studies attempted to understand the major limitations associated with cisplatin therapy, and so far, there is no definitive solution. As such, a more comprehensive molecular and genetic profiling of patients is needed to identify those individuals that can benefit from platinum therapy. Additionally, the treatment regimen can be improved by combining cisplatin with certain molecular targeted therapies to achieve a balance between tumour toxicity and tolerance mechanisms. In this review, we discuss the importance of various biological processes that contribute to the resistance of cisplatin and its derivatives. We aim to highlight the processes that can be modulated to suppress cisplatin resistance and provide an insight into the role of uptake transporters in enhancing drug efficacy.
Collapse
Affiliation(s)
- Reem Ali
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Qatar Foundation, Doha P.O. Box 34110, Qatar; (M.A.); (A.A.S.)
- Correspondence: (R.A.); (D.R.)
| | - Mustapha Aouida
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Qatar Foundation, Doha P.O. Box 34110, Qatar; (M.A.); (A.A.S.)
| | - Abdallah Alhaj Sulaiman
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Qatar Foundation, Doha P.O. Box 34110, Qatar; (M.A.); (A.A.S.)
| | - Srinivasan Madhusudan
- Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK;
| | - Dindial Ramotar
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Qatar Foundation, Doha P.O. Box 34110, Qatar; (M.A.); (A.A.S.)
- Correspondence: (R.A.); (D.R.)
| |
Collapse
|
22
|
Prieto-Garcia C, Hartmann O, Reissland M, Braun F, Bozkurt S, Pahor N, Fuss C, Schirbel A, Schülein-Völk C, Buchberger A, Calzado Canale MA, Rosenfeldt M, Dikic I, Münch C, Diefenbacher ME. USP28 enables oncogenic transformation of respiratory cells and its inhibition potentiates molecular therapy targeting mutant EGFR, BRAF and PI3K. Mol Oncol 2022; 16:3082-3106. [PMID: 35364627 PMCID: PMC9441007 DOI: 10.1002/1878-0261.13217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/04/2022] [Accepted: 03/29/2022] [Indexed: 11/23/2022] Open
Abstract
Oncogenic transformation of lung epithelial cells is a multistep process, frequently starting with the inactivation of tumour suppressors and subsequent development of activating mutations in proto‐oncogenes, such as members of the PI3K or MAPK families. Cells undergoing transformation have to adjust to changes, including altered metabolic requirements. This is achieved, in part, by modulating the protein abundance of transcription factors. Here, we report that the ubiquitin carboxyl‐terminal hydrolase 28 (USP28) enables oncogenic reprogramming by regulating the protein abundance of proto‐oncogenes such as c‐JUN, c‐MYC, NOTCH and ∆NP63 at early stages of malignant transformation. USP28 levels are increased in cancer compared with in normal cells due to a feed‐forward loop, driven by increased amounts of oncogenic transcription factors such as c‐MYC and c‐JUN. Irrespective of oncogenic driver, interference with USP28 abundance or activity suppresses growth and survival of transformed lung cells. Furthermore, inhibition of USP28 via a small‐molecule inhibitor resets the proteome of transformed cells towards a ‘premalignant’ state, and its inhibition synergizes with clinically established compounds used to target EGFRL858R‐, BRAFV600E‐ or PI3KH1047R‐driven tumour cells. Targeting USP28 protein abundance at an early stage via inhibition of its activity is therefore a feasible strategy for the treatment of early‐stage lung tumours, and the observed synergism with current standard‐of‐care inhibitors holds the potential for improved targeting of established tumours.
Collapse
Affiliation(s)
- Cristian Prieto-Garcia
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.,Mildred Scheel Early Career Center, Wuerzburg, Germany.,Molecular Signaling Group, Institute of Biochemistry II, Goethe University, Frankfurt, Germany
| | - Oliver Hartmann
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.,Mildred Scheel Early Career Center, Wuerzburg, Germany
| | - Michaela Reissland
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.,Mildred Scheel Early Career Center, Wuerzburg, Germany
| | - Fabian Braun
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.,Mildred Scheel Early Career Center, Wuerzburg, Germany
| | - Süleyman Bozkurt
- Protein quality control, Institute of Biochemistry II, Goethe University, Frankfurt, Germany
| | - Nikolett Pahor
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.,Mildred Scheel Early Career Center, Wuerzburg, Germany
| | - Carmina Fuss
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.,Mildred Scheel Early Career Center, Wuerzburg, Germany.,Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | | | | | - Marco A Calzado Canale
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Mathias Rosenfeldt
- Mildred Scheel Early Career Center, Wuerzburg, Germany.,Institut für Pathologie, Universitaetsklinikum Wuerzburg
| | - Ivan Dikic
- Molecular Signaling Group, Institute of Biochemistry II, Goethe University, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Germany
| | - Christian Münch
- Protein quality control, Institute of Biochemistry II, Goethe University, Frankfurt, Germany
| | - Markus E Diefenbacher
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.,Mildred Scheel Early Career Center, Wuerzburg, Germany
| |
Collapse
|
23
|
Pokorna Z, Vyslouzil J, Vojtesek B, Coates PJ. Identifying pathways regulating the oncogenic p53 family member ΔNp63 provides therapeutic avenues for squamous cell carcinoma. Cell Mol Biol Lett 2022; 27:18. [PMID: 35196980 PMCID: PMC8903560 DOI: 10.1186/s11658-022-00323-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/15/2022] [Indexed: 12/17/2022] Open
Abstract
Background ΔNp63 overexpression is a common event in squamous cell carcinoma (SCC) that contributes to tumorigenesis, making ΔNp63 a potential target for therapy. Methods We created inducible TP63-shRNA cells to study the effects of p63-depletion in SCC cell lines and non-malignant HaCaT keratinocytes. DNA damaging agents, growth factors, signaling pathway inhibitors, histone deacetylase inhibitors, and metabolism-modifying drugs were also investigated for their ability to influence ΔNp63 protein and mRNA levels. Results HaCaT keratinocytes, FaDu and SCC-25 cells express high levels of ΔNp63. HaCaT and FaDu inducible TP63-shRNA cells showed reduced proliferation after p63 depletion, with greater effects on FaDu than HaCaT cells, compatible with oncogene addiction in SCC. Genotoxic insults and histone deacetylase inhibitors variably reduced ΔNp63 levels in keratinocytes and SCC cells. Growth factors that regulate proliferation/survival of squamous cells (IGF-1, EGF, amphiregulin, KGF, and HGF) and PI3K, mTOR, MAPK/ERK or EGFR inhibitors showed lesser and inconsistent effects, with dual inhibition of PI3K and mTOR or EGFR inhibition selectively reducing ΔNp63 levels in HaCaT cells. In contrast, the antihyperlipidemic drug lovastatin selectively increased ΔNp63 in HaCaT cells. Conclusions These data confirm that ΔNp63-positive SCC cells require p63 for continued growth and provide proof of concept that p63 reduction is a therapeutic option for these tumors. Investigations of ΔNp63 regulation identified agent-specific and cell-specific pathways. In particular, dual inhibition of the PI3K and mTOR pathways reduced ΔNp63 more effectively than single pathway inhibition, and broad-spectrum histone deacetylase inhibitors showed a time-dependent biphasic response, with high level downregulation at the transcriptional level within 24 h. In addition to furthering our understanding of ΔNp63 regulation in squamous cells, these data identify novel drug combinations that may be useful for p63-based therapy of SCC. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00323-x.
Collapse
Affiliation(s)
- Zuzana Pokorna
- Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Jan Vyslouzil
- Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Borivoj Vojtesek
- Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Philip J Coates
- Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic.
| |
Collapse
|