87 results on '"Kuntian Luo"'
Search Results
2. Reciprocal regulation of RIG-I and XRCC4 connects DNA repair with RIG-I immune signaling
- Author
-
Guijie Guo, Ming Gao, Xiaochen Gao, Bibo Zhu, Jinzhou Huang, Xinyi Tu, Wootae Kim, Fei Zhao, Qin Zhou, Shouhai Zhu, Zheming Wu, Yuanliang Yan, Yong Zhang, Xiangyu Zeng, Qian Zhu, Ping Yin, Kuntian Luo, Jie Sun, Min Deng, and Zhenkun Lou
- Subjects
Science - Abstract
The RNA-sensing pathway has been associated with type I interferon (IFN) production induced by DNA damaging agents. Here the authors reveal that RIG-I, a cytosolic RNA sensor that recognizes RNA virus and initiates IFN signaling, is recruited to double-stranded breaks and suppresses non-homologous end joining.
- Published
- 2021
- Full Text
- View/download PDF
3. USP52 regulates DNA end resection and chemosensitivity through removing inhibitory ubiquitination from CtIP
- Author
-
Ming Gao, Guijie Guo, Jinzhou Huang, Jake A. Kloeber, Fei Zhao, Min Deng, Xinyi Tu, Wootae Kim, Qin Zhou, Chao Zhang, Ping Yin, Kuntian Luo, and Zhenkun Lou
- Subjects
Science - Abstract
C-terminal binding protein (CtBP) interacting protein (CtIP) is a fundamental factor for the initiation of DNA end resection to initiate DNA repair. Here the authors reveal mechanistic insights into the regulation of CtIP via the deubiquitinase USP52.
- Published
- 2020
- Full Text
- View/download PDF
4. The bromodomain containing protein BRD-9 orchestrates RAD51–RAD54 complex formation and regulates homologous recombination-mediated repair
- Author
-
Qin Zhou, Jinzhou Huang, Chao Zhang, Fei Zhao, Wootae Kim, Xinyi Tu, Yong Zhang, Somaira Nowsheen, Qian Zhu, Min Deng, Yuping Chen, Bo Qin, Kuntian Luo, Baohua Liu, Zhenkun Lou, Robert W. Mutter, and Jian Yuan
- Subjects
Science - Abstract
The bromodomain containing protein BRD9 has been reported to regulate chromatin remodeling and transcription. Here the authors reveal a role for BRD9 in homologous recombination by facilitating RAD51–RAD54 interaction.
- Published
- 2020
- Full Text
- View/download PDF
5. SARS-CoV-2 non-structural protein 13 (nsp13) hijacks host deubiquitinase USP13 and counteracts host antiviral immune response
- Author
-
Guijie Guo, Ming Gao, Xiaochen Gao, Bibo Zhu, Jinzhou Huang, Kuntian Luo, Yong Zhang, Jie Sun, Min Deng, and Zhenkun Lou
- Subjects
Medicine ,Biology (General) ,QH301-705.5 - Published
- 2021
- Full Text
- View/download PDF
6. Author Correction: The bromodomain containing protein BRD-9 orchestrates RAD51–RAD54 complex formation and regulates homologous recombination-mediated repair
- Author
-
Qin Zhou, Jinzhou Huang, Chao Zhang, Fei Zhao, Wootae Kim, Xinyi Tu, Yong Zhang, Somaira Nowsheen, Qian Zhu, Min Deng, Yuping Chen, Bo Qin, Kuntian Luo, Baohua Liu, Zhenkun Lou, Robert W. Mutter, and Jian Yuan
- Subjects
Science - Published
- 2022
- Full Text
- View/download PDF
7. ZFP161 regulates replication fork stability and maintenance of genomic stability by recruiting the ATR/ATRIP complex
- Author
-
Wootae Kim, Fei Zhao, Rentian Wu, Sisi Qin, Somaira Nowsheen, Jinzhou Huang, Qin Zhou, Yuping Chen, Min Deng, Guijie Guo, Kuntian Luo, Zhenkun Lou, and Jian Yuan
- Subjects
Science - Abstract
The ATR pathway is active during DNA replication stress to maintain genome stability. Here the authors reveal the role of the zinc finger containing protein 161 (ZFP161) to facilitate replication fork stability by acting as a scaffold to facilitate the interaction between RPA and ATR/ATRIP.
- Published
- 2019
- Full Text
- View/download PDF
8. LRRK2 inhibition potentiates PARP inhibitor cytotoxicity through inhibiting homologous recombination‐mediated DNA double strand break repair
- Author
-
Lifeng Chen, Jing Hou, Xiangyu Zeng, Qiang Guo, Min Deng, Jake A Kloeber, Xinyi Tu, Fei Zhao, Zheming Wu, Jinzhou Huang, Kuntian Luo, Wootae Kim, and Zhenkun Lou
- Subjects
HR ,LRRK2 inhibitor ,PARP inhibitor ,Rad51 ,Medicine (General) ,R5-920 - Abstract
Abstract PARP inhibitors induce DNA lesions, the repair of which are highly dependent on homologous recombination (HR), and preferentially kill HR‐ deficient cancers. However, cancer cells have developed several mechanisms to transform HR and confer drug resistance to PARP inhibition. Therefore, there is a great clinical interest in exploring new therapies that induce HR deficiency (HRD), thereby sensitizing cancer cells to PARP inhibitors. Here, we found that GSK2578215A, a high‐selective and effective leucine‐rich repeat kinase 2 (LRRK2) inhibitor, or LRRK2 depletion suppresses HR preventing the recruitment of RAD51 to DNA damage sites through disruption of the interaction of RAD51 and BRCA2. Moreover, LRRK2 inhibition or depletion increases the susceptibility of ovarian cancer cells to Olaparib in vitro and in vivo. In clinical specimens, LRRK2 high expression is high related with advanced clinical characteristics and poor survival of ovarian cancer patients. All these findings indicate ovarian cancers expressing high levels of LRRK2 are more resistant to treatment potentially through promoting HR. Furthermore, combination treatment with an LRRK2 and PARP inhibitor may be a novel strategy to improve the effectiveness of LRRK2 expression ovarian cancers.
- Published
- 2021
- Full Text
- View/download PDF
9. ZNF506-dependent positive feedback loop regulates H2AX signaling after DNA damage
- Author
-
Somaira Nowsheen, Khaled Aziz, Kuntian Luo, Min Deng, Bo Qin, Jian Yuan, Karthik B. Jeganathan, Jia Yu, Henan Zhang, Wei Ding, Jan M. van Deursen, and Zhenkun Lou
- Subjects
Science - Abstract
Following double-strand break a cascade of events leads to the recruitment of repair factors to damaged sites. Here the authors identify ZNF506 as a key factor that mediates post-translational modification changes in H2AX affecting the DNA damage response.
- Published
- 2018
- Full Text
- View/download PDF
10. USP13 regulates the RAP80-BRCA1 complex dependent DNA damage response
- Author
-
Yunhui Li, Kuntian Luo, Yujiao Yin, Chenming Wu, Min Deng, Lei Li, Yuping Chen, Somaira Nowsheen, Zhenkun Lou, and Jian Yuan
- Subjects
Science - Abstract
RAP80 helps to recruit BRCA1 to double-strand breaks, facilitating DNA damage responses. Here the authors report that phosphorylated USP13 deubiquitinates RAP80 after DNA damage, prompting recruitment to the break site.
- Published
- 2017
- Full Text
- View/download PDF
11. CDK4/6-dependent activation of DUB3 regulates cancer metastasis through SNAIL1
- Author
-
Tongzheng Liu, Jia Yu, Min Deng, Yujiao Yin, Haoxing Zhang, Kuntian Luo, Bo Qin, Yunhui Li, Chenming Wu, Tao Ren, Yang Han, Peng Yin, JungJin Kim, SeungBaek Lee, Jing Lin, Lizhi Zhang, Jun Zhang, Somaira Nowsheen, Liewei Wang, Judy Boughey, Matthew P. Goetz, Jian Yuan, and Zhenkun Lou
- Subjects
Science - Abstract
Overexpression of SNAIL confers tumour cells with cancer stem-like characteristics associated with tumour progression. Here the authors show that inhibition of CDK4/6 blocks tumour metastasis in triple negative breast cancer by targeting DUB3 which in turns deubiquitinates and stabilises SNAIL1.
- Published
- 2017
- Full Text
- View/download PDF
12. Supplementary Figure S6 from MET Amplification Attenuates Lung Tumor Response to Immunotherapy by Inhibiting STING
- Author
-
Zhenkun Lou, Guoliang Pi, Junqiu Yue, Sheng Hu, Lifeng Chen, Qian Zhu, Yu Shi, Jing Hou, Wootae Kim, Min Deng, Kuntian Luo, Ping Yin, Mingwei Wang, Kai Zhang, Li Liu, Jinzhou Huang, Fei Zhao, Qin Zhou, Guijie Guo, Yueyu Cao, Jake A. Kloeber, Qiang Guo, Chao Zhang, Wenzhuan Xie, Ting Wei, Xinyi Tu, Bin Yang, Shuang Dong, Manxiang Wang, Xiangyu Zeng, Qifan Yang, and Yong Zhang
- Abstract
Effects of MET and STING signals on MET OE tumor after the intervention of MET inhibitor in vivo
- Published
- 2023
13. Supplementary Methods from MET Amplification Attenuates Lung Tumor Response to Immunotherapy by Inhibiting STING
- Author
-
Zhenkun Lou, Guoliang Pi, Junqiu Yue, Sheng Hu, Lifeng Chen, Qian Zhu, Yu Shi, Jing Hou, Wootae Kim, Min Deng, Kuntian Luo, Ping Yin, Mingwei Wang, Kai Zhang, Li Liu, Jinzhou Huang, Fei Zhao, Qin Zhou, Guijie Guo, Yueyu Cao, Jake A. Kloeber, Qiang Guo, Chao Zhang, Wenzhuan Xie, Ting Wei, Xinyi Tu, Bin Yang, Shuang Dong, Manxiang Wang, Xiangyu Zeng, Qifan Yang, and Yong Zhang
- Abstract
Supplementary Methods
- Published
- 2023
14. Table S1 from MET Amplification Attenuates Lung Tumor Response to Immunotherapy by Inhibiting STING
- Author
-
Zhenkun Lou, Guoliang Pi, Junqiu Yue, Sheng Hu, Lifeng Chen, Qian Zhu, Yu Shi, Jing Hou, Wootae Kim, Min Deng, Kuntian Luo, Ping Yin, Mingwei Wang, Kai Zhang, Li Liu, Jinzhou Huang, Fei Zhao, Qin Zhou, Guijie Guo, Yueyu Cao, Jake A. Kloeber, Qiang Guo, Chao Zhang, Wenzhuan Xie, Ting Wei, Xinyi Tu, Bin Yang, Shuang Dong, Manxiang Wang, Xiangyu Zeng, Qifan Yang, and Yong Zhang
- Abstract
Baseline characteristics of patients
- Published
- 2023
15. Data from MET Amplification Attenuates Lung Tumor Response to Immunotherapy by Inhibiting STING
- Author
-
Zhenkun Lou, Guoliang Pi, Junqiu Yue, Sheng Hu, Lifeng Chen, Qian Zhu, Yu Shi, Jing Hou, Wootae Kim, Min Deng, Kuntian Luo, Ping Yin, Mingwei Wang, Kai Zhang, Li Liu, Jinzhou Huang, Fei Zhao, Qin Zhou, Guijie Guo, Yueyu Cao, Jake A. Kloeber, Qiang Guo, Chao Zhang, Wenzhuan Xie, Ting Wei, Xinyi Tu, Bin Yang, Shuang Dong, Manxiang Wang, Xiangyu Zeng, Qifan Yang, and Yong Zhang
- Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer therapy. However, the response of patients to ICB is difficult to predict. Here, we examined 81 patients with lung cancer under ICB treatment and found that patients with MET amplification were resistant to ICB and had a poor progression-free survival. Tumors with MET amplifications had significantly decreased STING levels and antitumor T-cell infiltration. Furthermore, we performed deep single-cell RNA sequencing on more than 20,000 single immune cells and identified an immunosuppressive signature with increased subsets of XIST- and CD96-positive exhausted natural killer (NK) cells and decreased CD8+ T-cell and NK-cell populations in patients with MET amplification. Mechanistically, we found that oncogenic MET signaling induces phosphorylation of UPF1 and downregulates tumor cell STING expression via modulation of the 3′-UTR length of STING by UPF1. Decreased efficiency of ICB by MET amplification can be overcome by inhibiting MET.Significance:We suggest that the combination of MET inhibitor together with ICB will overcome ICB resistance induced by MET amplification. Our report reveals much-needed information that will benefit the treatment of patients with primary MET amplification or EGFR–tyrosine kinase inhibitor resistant-related MET amplification.This article is highlighted in the In This Issue feature, p. 2659
- Published
- 2023
16. RNF19A-mediated ubiquitination of BARD1 prevents BRCA1/BARD1-dependent homologous recombination
- Author
-
Yong Zhang, Jiayi Chen, Qian Zhu, Min Deng, Peiqiang Yi, Hongyang Huang, Xinyi Tu, Jake A. Kloeber, Huan Li, Ming Gao, Guijie Guo, Yuping Chen, Jian Yuan, Jinzhou Huang, Zhenkun Lou, Kuntian Luo, Ping Yin, and Jun Su
- Subjects
Ubiquitylation ,endocrine system diseases ,Carcinogenesis ,DNA repair ,Ubiquitin-Protein Ligases ,Science ,General Physics and Astronomy ,Breast Neoplasms ,Poly(ADP-ribose) Polymerase Inhibitors ,Article ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,Breast cancer ,Ubiquitin ,BARD1 ,Humans ,Homologous recombination ,Nuclear export signal ,skin and connective tissue diseases ,Polymerase ,Multidisciplinary ,biology ,BRCA1 Protein ,Tumor Suppressor Proteins ,Ubiquitination ,General Chemistry ,Cell biology ,chemistry ,Cancer cell ,biology.protein ,Female ,Protein Multimerization ,RING Finger Domains ,DNA ,DNA Damage ,Protein Binding - Abstract
BRCA1-BARD1 heterodimers act in multiple steps during homologous recombination (HR) to ensure the prompt repair of DNA double strand breaks. Dysfunction of the BRCA1 pathway enhances the therapeutic efficiency of poly-(ADP-ribose) polymerase inhibitors (PARPi) in cancers, but the molecular mechanisms underlying this sensitization to PARPi are not fully understood. Here, we show that cancer cell sensitivity to PARPi is promoted by the ring between ring fingers (RBR) protein RNF19A. We demonstrate that RNF19A suppresses HR by ubiquitinating BARD1, which leads to dissociation of BRCA1-BARD1 complex and exposure of a nuclear export sequence in BARD1 that is otherwise masked by BRCA1, resulting in the export of BARD1 to the cytoplasm. We provide evidence that high RNF19A expression in breast cancer compromises HR and increases sensitivity to PARPi. We propose that RNF19A modulates the cancer cell response to PARPi by negatively regulating the BRCA1-BARD1 complex and inhibiting HR-mediated DNA repair., BRCA1 dysfunction sensitizes cancer cells to PARP inhibitors (PARPi) but the underlying mechanism is unclear. Here, the authors identify RNF19A as a determinant of PARPi sensitivity, showing that RNF19A ubiquitinates BARD1, negatively regulates the BRCA1-BARD1 complex, and inhibits homologous recombination.
- Published
- 2021
17. ASTE1 promotes shieldin-complex-mediated DNA repair by attenuating end resection
- Author
-
Ming Gao, Qin Zhou, Shih-Hsun Chen, Yong Zhang, Qi Hu, Yilun Liu, Ping Yin, Bo Qin, Sisi Qin, Somaira Nowsheen, Jian Yuan, Chao Zhang, Xinyi Tu, Zhenkun Lou, Jinzhou Huang, Kuntian Luo, Yuping Chen, Fei Zhao, Guijie Guo, Jake A. Kloeber, Huanyao Gao, Wootae Kim, Min Deng, and Chao Liu
- Subjects
Genome instability ,biology ,Chemistry ,DNA repair ,DNA damage ,Cell Biology ,Immunoglobulin Class Switch Recombination ,Cell biology ,DNA End-Joining Repair ,chemistry.chemical_compound ,biology.protein ,Homologous recombination ,Polymerase ,DNA - Abstract
The shieldin complex functions as the downstream effector of 53BP1-RIF1 to promote DNA double-strand break end-joining by restricting end resection. The SHLD2 subunit binds to single-stranded DNA ends and blocks end resection through OB-fold domains. Besides blocking end resection, it is unclear how the shieldin complex processes SHLD2-bound single-stranded DNA and promotes non-homologous end-joining. Here, we identify a downstream effector of the shieldin complex, ASTE1, as a structure-specific DNA endonuclease that specifically cleaves single-stranded DNA and 3' overhang DNA. ASTE1 localizes to DNA damage sites in a shieldin-dependent manner. Loss of ASTE1 impairs non-homologous end-joining, leads to hyper-resection and causes defective immunoglobulin class switch recombination. ASTE1 deficiency also causes resistance to poly(ADP-ribose) polymerase inhibitors in BRCA1-deficient cells owing to restoration of homologous recombination. These findings suggest that ASTE1-mediated 3' single-stranded DNA end cleavage contributes to the control of DSB repair choice by 53BP1, RIF1 and shieldin.
- Published
- 2021
18. DOCK7 protects against replication stress by promoting RPA stability on chromatin
- Author
-
Qian Zhu, Ming Gao, John Weroha, Ping Yin, Xinyi Tu, Wootae Kim, Min Deng, Jiaqi Liu, Jinzhou Huang, Daniel D. Billadeau, Qin Zhou, Hyoungjun Ham, Yuanliang Yan, Guijie Guo, Fei Zhao, Chao Zhang, Zhijie Xu, Zhenkun Lou, Xiaonan Hou, and Kuntian Luo
- Subjects
DNA Replication ,rac1 GTP-Binding Protein ,DNA Repair ,AcademicSubjects/SCI00010 ,DNA repair ,Regulator ,Mice, Nude ,Genome Integrity, Repair and Replication ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Stress, Physiological ,Cell Line, Tumor ,Replication Protein A ,Genetics ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Phosphorylation ,cdc42 GTP-Binding Protein ,030304 developmental biology ,Ovarian Neoplasms ,0303 health sciences ,GTPase-Activating Proteins ,DNA replication ,DNA Replication Fork ,Chromatin ,MDC1 ,Cell biology ,HEK293 Cells ,p21-Activated Kinases ,Cdc42 GTP-Binding Protein ,030220 oncology & carcinogenesis ,Proteolysis ,Cancer cell ,Female ,Signal Transduction - Abstract
RPA is a critical factor for DNA replication and replication stress response. Surprisingly, we found that chromatin RPA stability is tightly regulated. We report that the GDP/GTP exchange factor DOCK7 acts as a critical replication stress regulator to promote RPA stability on chromatin. DOCK7 is phosphorylated by ATR and then recruited by MDC1 to the chromatin and replication fork during replication stress. DOCK7-mediated Rac1/Cdc42 activation leads to the activation of PAK1, which subsequently phosphorylates RPA1 at S135 and T180 to stabilize chromatin-loaded RPA1 and ensure proper replication stress response. Moreover, DOCK7 is overexpressed in ovarian cancer and depleting DOCK7 sensitizes cancer cells to camptothecin. Taken together, our results highlight a novel role for DOCK7 in regulation of the replication stress response and highlight potential therapeutic targets to overcome chemoresistance in cancer.
- Published
- 2021
19. Correction to: USP20 positively regulates tumorigenesis and chemoresistance through β-catenin stabilization
- Author
-
Chenming Wu, Kuntian Luo, Fei Zhao, Ping Yin, Ying Song, Min Deng, Jinzhou Huang, Yuping Chen, Lei Li, SeungBaek Lee, JungJin Kim, Qin Zhou, Xinyi Tu, Somaira Nowsheen, Qifeng Luo, Xiumei Gao, Zhenkun Lou, Zhongmin Liu, and Jian Yuan
- Subjects
Cell Biology ,Molecular Biology - Published
- 2022
20. USP52 regulates DNA end resection and chemosensitivity through removing inhibitory ubiquitination from CtIP
- Author
-
Chao Zhang, Qin Zhou, Wootae Kim, Guijie Guo, Min Deng, Ming Gao, Kuntian Luo, Fei Zhao, Jake A. Kloeber, Jinzhou Huang, Xinyi Tu, Ping Yin, and Zhenkun Lou
- Subjects
0301 basic medicine ,Ubiquitylation ,DNA repair ,DNA damage ,Science ,Poly ADP ribose polymerase ,Mice, Nude ,General Physics and Astronomy ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Ubiquitin ,Animals ,Humans ,Phosphorylation ,Homologous Recombination ,lcsh:Science ,Endodeoxyribonucleases ,Multidisciplinary ,biology ,HEK 293 cells ,Ubiquitination ,DNA ,General Chemistry ,Xenograft Model Antitumor Assays ,Cell biology ,DNA-Binding Proteins ,HEK293 Cells ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Exoribonucleases ,biology.protein ,Female ,lcsh:Q ,Homologous recombination ,Protein Processing, Post-Translational ,DNA Damage ,Protein Binding ,Post-translational modifications - Abstract
Human C-terminal binding protein (CtBP)–interacting protein (CtIP) is a central regulator to initiate DNA end resection and homologous recombination (HR). Several studies have shown that post-translational modifications control the activity or expression of CtIP. However, it remains unclear whether and how cells restrain CtIP activity in unstressed cells and activate CtIP when needed. Here, we identify that USP52 directly interacts with and deubiquitinates CtIP, thereby promoting DNA end resection and HR. Mechanistically, USP52 removes the ubiquitination of CtIP to facilitate the phosphorylation and activation of CtIP at Thr-847. In addition, USP52 is phosphorylated by ATM at Ser-1003 after DNA damage, which enhances the catalytic activity of USP52. Furthermore, depletion of USP52 sensitizes cells to PARP inhibition in a CtIP-dependent manner in vitro and in vivo. Collectively, our findings reveal the key role of USP52 and the regulatory complexity of CtIP deubiquitination in DNA repair., C-terminal binding protein (CtBP) interacting protein (CtIP) is a fundamental factor for the initiation of DNA end resection to initiate DNA repair. Here the authors reveal mechanistic insights into the regulation of CtIP via the deubiquitinase USP52.
- Published
- 2020
21. SUMOylation of HNRNPA2B1 modulates RPA dynamics during unperturbed replication and genotoxic stress responses
- Author
-
Shouhai Zhu, Jing Hou, Huanyao Gao, Qi Hu, Jake A. Kloeber, Jinzhou Huang, Fei Zhao, Qin Zhou, Kuntian Luo, Zheming Wu, Xinyi Tu, Ping Yin, and Zhenkun Lou
- Subjects
Cell Biology ,Molecular Biology - Published
- 2023
22. METTL16 antagonizes MRE11-mediated DNA end resection and confers synthetic lethality to PARP inhibition in pancreatic ductal adenocarcinoma
- Author
-
Xiangyu Zeng, Fei Zhao, Gaofeng Cui, Yong Zhang, Rajashree A. Deshpande, Yuping Chen, Min Deng, Jake A. Kloeber, Yu Shi, Qin Zhou, Chao Zhang, Jing Hou, Wootae Kim, Xinyi Tu, Yuanliang Yan, Zhijie Xu, Lifeng Chen, Huanyao Gao, Guijie Guo, Jiaqi Liu, Qian Zhu, Yueyu Cao, Jinzhou Huang, Zheming Wu, Shouhai Zhu, Ping Yin, Kuntian Luo, Georges Mer, Tanya T. Paull, Jian Yuan, Kaixiong Tao, and Zhenkun Lou
- Subjects
Exonucleases ,Cancer Research ,Adenosine Diphosphate Ribose ,MRE11 Homologue Protein ,DNA ,Methyltransferases ,Poly(ADP-ribose) Polymerase Inhibitors ,Pancreatic Neoplasms ,Oncology ,Humans ,RNA ,Poly(ADP-ribose) Polymerases ,Synthetic Lethal Mutations ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers. Characterization of genetic alterations will improve our understanding and therapies for this disease. Here, we report that PDAC with elevated expression of METTL16, one of the 'writers' of RNA N
- Published
- 2021
23. Reciprocal regulation of RIG-I and XRCC4 connects DNA repair with RIG-I immune signaling
- Author
-
Yong Zhang, Fei Zhao, Ping Yin, Xiaochen Gao, Qin Zhou, Ming Gao, Jinzhou Huang, Bibo Zhu, Zheming Wu, Yuanliang Yan, Zhenkun Lou, Xiangyu Zeng, Kuntian Luo, Wootae Kim, Min Deng, Shouhai Zhu, Guijie Guo, Xinyi Tu, Jie Sun, and Qian Zhu
- Subjects
0301 basic medicine ,Cell biology ,DNA End-Joining Repair ,DNA Repair ,DNA repair ,Science ,viruses ,General Physics and Astronomy ,chemical and pharmacologic phenomena ,Virus Replication ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Retrovirus ,Interferon ,Radiation, Ionizing ,medicine ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Receptors, Immunologic ,Virus Integration ,Cancer ,Multidisciplinary ,biology ,Genome, Human ,RIG-I ,virus diseases ,RNA ,Dose-Response Relationship, Radiation ,RNA virus ,General Chemistry ,biochemical phenomena, metabolism, and nutrition ,DNA repair protein XRCC4 ,biology.organism_classification ,DNA-Binding Proteins ,HEK293 Cells ,Retroviridae ,030104 developmental biology ,A549 Cells ,DEAD Box Protein 58 ,biological phenomena, cell phenomena, and immunity ,030217 neurology & neurosurgery ,Signal Transduction ,medicine.drug - Abstract
The RNA-sensing pathway contributes to type I interferon (IFN) production induced by DNA damaging agents. However, the potential involvement of RNA sensors in DNA repair is unknown. Here, we found that retinoic acid-inducible gene I (RIG-I), a key cytosolic RNA sensor that recognizes RNA virus and initiates the MAVS-IRF3-type I IFN signaling cascade, is recruited to double-stranded breaks (DSBs) and suppresses non-homologous end joining (NHEJ). Mechanistically, RIG-I interacts with XRCC4, and the RIG-I/XRCC4 interaction impedes the formation of XRCC4/LIG4/XLF complex at DSBs. High expression of RIG-I compromises DNA repair and sensitizes cancer cells to irradiation treatment. In contrast, depletion of RIG-I renders cells resistant to irradiation in vitro and in vivo. In addition, this mechanism suggests a protective role of RIG-I in hindering retrovirus integration into the host genome by suppressing the NHEJ pathway. Reciprocally, XRCC4, while suppressed for its DNA repair function, has a critical role in RIG-I immune signaling through RIG-I interaction. XRCC4 promotes RIG-I signaling by enhancing oligomerization and ubiquitination of RIG-I, thereby suppressing RNA virus replication in host cells. In vivo, silencing XRCC4 in mouse lung promotes influenza virus replication in mice and these mice display faster body weight loss, poorer survival, and a greater degree of lung injury caused by influenza virus infection. This reciprocal regulation of RIG-I and XRCC4 reveals a new function of RIG-I in suppressing DNA repair and virus integration into the host genome, and meanwhile endues XRCC4 with a crucial role in potentiating innate immune response, thereby helping host to prevail in the battle against virus., The RNA-sensing pathway has been associated with type I interferon (IFN) production induced by DNA damaging agents. Here the authors reveal that RIG-I, a cytosolic RNA sensor that recognizes RNA virus and initiates IFN signaling, is recruited to double-stranded breaks and suppresses non-homologous end joining.
- Published
- 2021
24. LRRK2 inhibition potentiates PARP inhibitor cytotoxicity through inhibiting homologous recombination‐mediated DNA double strand break repair
- Author
-
Jinzhou Huang, Guo Qiang, Wootae Kim, Xiangyu Zeng, Min Deng, Jing Hou, Kuntian Luo, Lifeng Chen, Zheming Wu, Fei Zhao, Zhenkun Lou, Jake A. Kloeber, and Xinyi Tu
- Subjects
0301 basic medicine ,DNA damage ,Poly ADP ribose polymerase ,RAD51 ,Mice, Nude ,Medicine (miscellaneous) ,Antineoplastic Agents ,Poly(ADP-ribose) Polymerase Inhibitors ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,LRRK2 inhibitor ,Olaparib ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,HR ,medicine ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Research Articles ,Ovarian Neoplasms ,lcsh:R5-920 ,Chemistry ,Recombinational DNA Repair ,Drug Synergism ,medicine.disease ,nervous system diseases ,Disease Models, Animal ,PARP inhibitor ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Rad51 ,Cancer research ,Molecular Medicine ,Female ,lcsh:Medicine (General) ,Homologous recombination ,Ovarian cancer ,Research Article - Abstract
PARP inhibitors induce DNA lesions, the repair of which are highly dependent on homologous recombination (HR), and preferentially kill HR‐ deficient cancers. However, cancer cells have developed several mechanisms to transform HR and confer drug resistance to PARP inhibition. Therefore, there is a great clinical interest in exploring new therapies that induce HR deficiency (HRD), thereby sensitizing cancer cells to PARP inhibitors. Here, we found that GSK2578215A, a high‐selective and effective leucine‐rich repeat kinase 2 (LRRK2) inhibitor, or LRRK2 depletion suppresses HR preventing the recruitment of RAD51 to DNA damage sites through disruption of the interaction of RAD51 and BRCA2. Moreover, LRRK2 inhibition or depletion increases the susceptibility of ovarian cancer cells to Olaparib in vitro and in vivo. In clinical specimens, LRRK2 high expression is high related with advanced clinical characteristics and poor survival of ovarian cancer patients. All these findings indicate ovarian cancers expressing high levels of LRRK2 are more resistant to treatment potentially through promoting HR. Furthermore, combination treatment with an LRRK2 and PARP inhibitor may be a novel strategy to improve the effectiveness of LRRK2 expression ovarian cancers., LRRK2 inhibitor GSK2578215A suppresses HR and sensitizes ovarian cancer cells to PARP inhibitor. LRRK2 inhibition impedes the recruitment of RAD51 by disrupting the interaction of RAD51 and BRCA2. High expression of LRRK2 is associated with poor survival of ovarian cancer patients.
- Published
- 2021
25. The deubiquitinase USP36 Regulates DNA replication stress and confers therapeutic resistance through PrimPol stabilization
- Author
-
Wootae Kim, Kuntian Luo, Xiangyu Zeng, Zhenkun Lou, Jinzhou Huang, Qian Zhu, Ming Gao, Yuanliang Yan, Fei Zhao, Jake A. Kloeber, Zhijie Xu, and Guijie Guo
- Subjects
DNA Replication ,DNA damage ,AcademicSubjects/SCI00010 ,DNA Primase ,DNA-Directed DNA Polymerase ,Biology ,Genome Integrity, Repair and Replication ,Piperazines ,Deubiquitinating enzyme ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Ubiquitin ,Downregulation and upregulation ,Cell Line, Tumor ,Genetics ,Humans ,Polyubiquitin ,Polymerase ,030304 developmental biology ,Regulation of gene expression ,Ovarian Neoplasms ,0303 health sciences ,Deubiquitinating Enzymes ,Protein Stability ,DNA replication ,Prognosis ,Multifunctional Enzymes ,Cell biology ,Gene Expression Regulation, Neoplastic ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Proteolysis ,biology.protein ,Phthalazines ,Female ,Cisplatin ,Ubiquitin Thiolesterase ,DNA Damage - Abstract
PrimPol has been recently identified as a DNA damage tolerant polymerase that plays an important role in replication stress response. However, the regulatory mechanisms of PrimPol are not well defined. In this study, we identify that the deubiquitinase USP36 interferes with degradation of PrimPol to regulate the replication stress response. Mechanistically, USP36 is deubiquitinated following DNA replication stress, which in turn facilitates its upregulation and interaction with PrimPol. USP36 deubiquitinates K29-linked polyubiquitination of PrimPol and increases its protein stability. Depletion of USP36 results in replication stress-related defects and elevates cell sensitivity to DNA-damage agents, such as cisplatin and olaparib. Moreover, USP36 expression positively correlates with the level of PrimPol protein and poor prognosis in patient samples. These findings indicate that the regulation of PrimPol K29-linked ubiquitination by USP36 plays a critical role in DNA replication stress and chemotherapy response.
- Published
- 2020
26. MET Amplification Attenuates Lung Tumor Response to Immunotherapy by Inhibiting STING
- Author
-
Guijie Guo, Yu Shi, Fei Zhao, Jake A. Kloeber, Yueyu Cao, Xiangyu Zeng, Junqiu Yue, Li Liu, Kuntian Luo, Xinyi Tu, Yong Zhang, Ping Yin, Ting Wei, Guoliang Pi, Mingwei Wang, Zhenkun Lou, Jing Hou, Manxiang Wang, Qifan Yang, Chao Zhang, Sheng Hu, Wootae Kim, Jinzhou Huang, Min Deng, Qin Zhou, Kai Zhang, Shuang Dong, Lifeng Chen, Bin Yang, Qian Zhu, Guo Qiang, and Wenzhuan Xie
- Subjects
0301 basic medicine ,Lung Neoplasms ,medicine.medical_treatment ,CD8-Positive T-Lymphocytes ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Medicine ,Humans ,Lung cancer ,Kinase ,business.industry ,Gene Amplification ,Membrane Proteins ,Immunotherapy ,Oncogenes ,Proto-Oncogene Proteins c-met ,medicine.disease ,Immune checkpoint ,Blockade ,Killer Cells, Natural ,Sting ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,business ,CD8 - Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer therapy. However, the response of patients to ICB is difficult to predict. Here, we examined 81 patients with lung cancer under ICB treatment and found that patients with MET amplification were resistant to ICB and had a poor progression-free survival. Tumors with MET amplifications had significantly decreased STING levels and antitumor T-cell infiltration. Furthermore, we performed deep single-cell RNA sequencing on more than 20,000 single immune cells and identified an immunosuppressive signature with increased subsets of XIST- and CD96-positive exhausted natural killer (NK) cells and decreased CD8+ T-cell and NK-cell populations in patients with MET amplification. Mechanistically, we found that oncogenic MET signaling induces phosphorylation of UPF1 and downregulates tumor cell STING expression via modulation of the 3′-UTR length of STING by UPF1. Decreased efficiency of ICB by MET amplification can be overcome by inhibiting MET. Significance: We suggest that the combination of MET inhibitor together with ICB will overcome ICB resistance induced by MET amplification. Our report reveals much-needed information that will benefit the treatment of patients with primary MET amplification or EGFR–tyrosine kinase inhibitor resistant-related MET amplification. This article is highlighted in the In This Issue feature, p. 2659
- Published
- 2020
27. SARS-CoV-2 non-structural protein 13 (nsp13) hijacks host deubiquitinase USP13 and counteracts host antiviral immune response
- Author
-
Kuntian Luo, Bibo Zhu, Guijie Guo, Zhenkun Lou, Ming Gao, Min Deng, Xiaochen Gao, Jie Sun, Yong Zhang, and Jinzhou Huang
- Subjects
Cancer Research ,Ubiquitin-Specific Proteases ,Cell biology ,Letter ,Molecular biology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,lcsh:Medicine ,Viral Nonstructural Proteins ,Deubiquitinating enzyme ,Immune system ,Immunity ,Genetics ,Humans ,lcsh:QH301-705.5 ,biology ,Host (biology) ,SARS-CoV-2 ,lcsh:R ,HEK 293 cells ,Structural protein ,COVID-19 ,Virology ,Immunity, Innate ,HEK293 Cells ,lcsh:Biology (General) ,A549 Cells ,biology.protein - Published
- 2020
28. ASTE1 promotes shieldin-complex-mediated DNA repair by attenuating end resection
- Author
-
Fei, Zhao, Wootae, Kim, Huanyao, Gao, Chao, Liu, Yong, Zhang, Yuping, Chen, Min, Deng, Qin, Zhou, Jinzhou, Huang, Qi, Hu, Shih-Hsun, Chen, Somaira, Nowsheen, Jake A, Kloeber, Bo, Qin, Ping, Yin, Xinyi, Tu, Guijie, Guo, Sisi, Qin, Chao, Zhang, Ming, Gao, Kuntian, Luo, Yilun, Liu, Zhenkun, Lou, and Jian, Yuan
- Subjects
Male ,DNA End-Joining Repair ,Recombinant Fusion Proteins ,Proteins ,Cell Cycle Proteins ,DNA ,Immunoglobulin Class Switching ,Genomic Instability ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Mice ,HEK293 Cells ,Animals ,Deoxyribonuclease I ,Humans ,Female - Abstract
The shieldin complex functions as the downstream effector of 53BP1-RIF1 to promote DNA double-strand break end-joining by restricting end resection. The SHLD2 subunit binds to single-stranded DNA ends and blocks end resection through OB-fold domains. Besides blocking end resection, it is unclear how the shieldin complex processes SHLD2-bound single-stranded DNA and promotes non-homologous end-joining. Here, we identify a downstream effector of the shieldin complex, ASTE1, as a structure-specific DNA endonuclease that specifically cleaves single-stranded DNA and 3' overhang DNA. ASTE1 localizes to DNA damage sites in a shieldin-dependent manner. Loss of ASTE1 impairs non-homologous end-joining, leads to hyper-resection and causes defective immunoglobulin class switch recombination. ASTE1 deficiency also causes resistance to poly(ADP-ribose) polymerase inhibitors in BRCA1-deficient cells owing to restoration of homologous recombination. These findings suggest that ASTE1-mediated 3' single-stranded DNA end cleavage contributes to the control of DSB repair choice by 53BP1, RIF1 and shieldin.
- Published
- 2020
29. CHK2-FOXK axis promotes transcriptional control of autophagy programs
- Author
-
Yunhui Li, Kuntian Luo, Jiaqi Liu, Jinhuan Wu, Zhongmin Liu, Wootae Kim, Min Deng, Ping Yin, Chengming Wu, Qin Zhou, Somaira Nowsheen, Yi-Han Chen, Xinyi Tu, Yuping Chen, Guohe Geng, Fei Zhao, Guijie Guo, Guang Liang, Jian Yuan, Zhenkun Lou, Lei Li, Chao Zhang, Jinzhou Huang, and Xiumei Gao
- Subjects
animal structures ,DNA damage ,Hyperphosphorylation ,environment and public health ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,law ,Transcription (biology) ,Neoplasms ,Autophagy ,Transcriptional regulation ,Humans ,Phosphorylation ,Checkpoint Kinase 2 ,Research Articles ,Cancer ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Binding Sites ,Multidisciplinary ,Chemistry ,SciAdv r-articles ,Forkhead Transcription Factors ,Hep G2 Cells ,Cell Biology ,Cell biology ,Gene Expression Regulation, Neoplastic ,enzymes and coenzymes (carbohydrates) ,14-3-3 Proteins ,A549 Cells ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Suppressor ,biological phenomena, cell phenomena, and immunity ,Cisplatin ,DNA Damage ,Protein Binding ,Signal Transduction ,Research Article - Abstract
We clarify that DNA damage triggers autophagy by CHK2-FOXK axis, and misregulation of this pathway leads to chemoresistance., Autophagy is an evolutionarily conserved catabolic process, which plays a vital role in removing misfolded proteins and clearing damaged organelles to maintain internal environment homeostasis. Here, we uncovered the checkpoint kinase 2 (CHK2)–FOXK (FOXK1 and FOXK2) axis playing an important role in DNA damage–mediated autophagy at the transcriptional regulation layer. Mechanistically, following DNA damage, CHK2 phosphorylates FOXK and creates a 14-3-3γ binding site, which, in turn, traps FOXK proteins in the cytoplasm. Because FOXK functions as the transcription suppressor of ATGs, DNA damage–mediated FOXKs’ cytoplasmic trapping induces autophagy. In addition, we found that a cancer-derived FOXK mutation induces FOXK hyperphosphorylation and enhances autophagy, resulting in chemoresistance. Cotreatment with cisplatin and chloroquine overcomes the chemoresistance caused by FOXK mutation. Overall, our study highlights a mechanism whereby DNA damage triggers autophagy by increasing autophagy genes via CHK2-FOXK–mediated transcriptional control, and misregulation of this pathway contributes to chemoresistance.
- Published
- 2020
30. ZNF506-dependent positive feedback loop regulates H2AX signaling after DNA damage
- Author
-
Khaled Aziz, Kuntian Luo, Wei Ding, Zhenkun Lou, Jia Yu, Somaira Nowsheen, Bo Qin, Karthik B. Jeganathan, Jan M. van Deursen, Henan Zhang, Jian Yuan, and Min Deng
- Subjects
0301 basic medicine ,DNA Repair ,General Physics and Astronomy ,Cell Cycle Proteins ,law.invention ,Histones ,chemistry.chemical_compound ,Mice ,law ,Phosphorylation ,lcsh:Science ,Feedback, Physiological ,B-Lymphocytes ,Multidisciplinary ,Gene Expression Regulation, Leukemic ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Recombinant Proteins ,Cell biology ,Histone ,Recombinant DNA ,Signal transduction ,Signal Transduction ,DNA damage ,DNA repair ,Science ,Kruppel-Like Transcription Factors ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,Animals ,Humans ,Adaptor Proteins, Signal Transducing ,Osteoblasts ,Epithelial Cells ,General Chemistry ,Survival Analysis ,MDC1 ,Repressor Proteins ,030104 developmental biology ,HEK293 Cells ,chemistry ,Leukemia, Prolymphocytic, T-Cell ,biology.protein ,Trans-Activators ,lcsh:Q ,Protein Tyrosine Phosphatases ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,DNA ,DNA Damage - Abstract
Cells respond to cytotoxic DNA double-strand breaks by recruiting repair proteins to the damaged site. Phosphorylation of the histone variant H2AX at S139 and Y142 modulate its interaction with downstream DNA repair proteins and their recruitment to DNA lesions. Here we report ATM-dependent ZNF506 localization to the lesion through MDC1 following DNA damage. ZNF506, in turn, recruits the protein phosphatase EYA, resulting in dephosphorylation of H2AX at Y142, which further facilitates the recruitment of MDC1 and other downstream repair factors. Thus, ZNF506 regulates the early dynamic signaling in the DNA damage response (DDR) pathway and controls progressive downstream signal amplification. Cells lacking ZNF506 or harboring mutations found in cancer patient samples are more sensitive to radiation, offering a potential new therapeutic option for cancers with mutations in this pathway. Taken together, these results demonstrate how the DDR pathway is orchestrated by ZNF506 to maintain genomic integrity., Following double-strand break a cascade of events leads to the recruitment of repair factors to damaged sites. Here the authors identify ZNF506 as a key factor that mediates post-translational modification changes in H2AX affecting the DNA damage response.
- Published
- 2018
31. USP20 positively regulates tumorigenesis and chemoresistance through β-catenin stabilization
- Author
-
Qin Zhou, Jinzhou Huang, Xiumei Gao, Fei Zhao, Zhongmin Liu, Min Deng, Ping Yin, Lei Li, Zhenkun Lou, Seung Baek Lee, Jung Jin Kim, Somaira Nowsheen, Chenming Wu, Jian Yuan, Xinyi Tu, Yuping Chen, Kuntian Luo, Qifeng Luo, and Ying Song
- Subjects
0301 basic medicine ,Cell Survival ,Leupeptins ,Mice, Nude ,Antineoplastic Agents ,Biology ,medicine.disease_cause ,Article ,Mice ,03 medical and health sciences ,Cell Movement ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,RNA, Small Interfering ,Wnt Signaling Pathway ,Molecular Biology ,beta Catenin ,Tissue homeostasis ,HEK 293 cells ,Ubiquitination ,Wnt signaling pathway ,Correction ,Cell Biology ,Cell Transformation, Neoplastic ,HEK293 Cells ,030104 developmental biology ,Drug Resistance, Neoplasm ,Catenin ,Cancer cell ,Cancer research ,Female ,RNA Interference ,Signal transduction ,Carcinogenesis ,Ubiquitin Thiolesterase ,Deubiquitination - Abstract
β-catenin is a major transcriptional activator of the canonical Wnt/β-catenin signaling pathway. It is important for a series of biological processes including tissue homeostasis, and embryonic development and is involved in various human diseases. Elevated oncogenic activity of β-catenin is frequently observed in cancers, which contributes to survival, metastasis and chemo-resistance of cancer cells. However, the mechanism of β-catenin overexpression in cancers is not well defined. Here we demonstrate that the deubiquitination enzyme USP20 is a new regulator of the Wnt/β-catenin signaling pathway. Mechanistically, USP20 regulates the deubiquitination of β-catenin to control its stability, thereby inducing proliferation, invasion and migration of cancer cells. High expression of USP20 correlates with increased β-catenin protein level in multiple cancer cell lines and patient samples. Moreover, knockdown of USP20 increases β-catenin polyubiquitination, which enhances β-catenin turnover and cell sensitivity to chemotherapy. Collectively, our results establish the USP20-β-catenin axis as a critical regulatory mechanism of canonical Wnt/β-catenin signaling pathway with an important role in tumorigenesis and chemo response in human cancers.
- Published
- 2018
32. L3MBTL2 orchestrates ubiquitin signalling by dictating the sequential recruitment of RNF8 and RNF168 after DNA damage
- Author
-
Karthikbabu B Jeganathan, Yibin Deng, Kuntian Luo, Somaira Nowsheen, Zhenkun Lou, Bo Qin, Jan M. van Deursen, Asef Aziz, Khaled Aziz, Jian Yuan, Jia Yu, Wei Ding, Tongzheng Liu, Min Deng, and Henan Zhang
- Subjects
0301 basic medicine ,RNF168 ,DNA Repair ,DNA repair ,DNA damage ,Ubiquitin-Protein Ligases ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,DNA damage response ,RNF8 ,03 medical and health sciences ,chemistry.chemical_compound ,Ubiquitin ,ubiquitin ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Humans ,MDC1 ,DNA Breaks, Double-Stranded ,L3MBTL2 ,Phosphorylation ,E3 ligase ,Adaptor Proteins, Signal Transducing ,Osteosarcoma ,biology ,HEK 293 cells ,Ubiquitination ,Nuclear Proteins ,Signal transducing adaptor protein ,Cell Biology ,Microreview ,Cell biology ,Chromatin ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,Protein Transport ,HEK293 Cells ,030104 developmental biology ,chemistry ,DNA double strand break repair ,ATM ,Trans-Activators ,biology.protein ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,DNA ,Signal Transduction ,Transcription Factors - Abstract
Genomic stress leads to various forms of DNA damage, of which DNA double strand breaks (DSBs) are the most lethal. An army of signaling molecules is called to action as soon as these DNA breaks are detected. Various protein modifications, such as phosphorylation and ubiquitination, are an integral part of the reaction. While phosphorylation activates various proteins, ubiquitin (Ub) adducts typically act as docking sites for DNA repair factors. The response to DNA DSB starts with the protein kinase ATM phosphorylating various substrates including MDC1 and histone H2AX. This mediator protein, MDC1, then recognizes phosphorylated histone H2AX and amplifies the damage response. The E3 ligase, RNF8, recognizes and binds to phosphorylated MDC1. RNF8 then modifies an unknown protein to call a second ubiquitin ligase, RNF168, into action. It has been recognized that these two ubiquitin ligases are recruited sequentially but there is an unknown linker protein between them. These two ubiquitin ligases are crucial to the formation of DSB-associated ubiquitin conjugates and, as a result, there has been long standing interest in the field in identifying the link between the two factors. In this paper we identify lethal(3) malignant brain tumor like 2 (L3MBTL2) as the substrate of RNF8 (Nowsheen S, et al. Nat Cell Biol 20:455-464, 2018). We report that ATM-mediated phosphorylation of the polycomb group like protein L3MBTL2 and subsequent interaction with MDC1 brings it to the vicinity of the DNA lesion. RNF8 acts upon this phosphorylated L3MBTL2 and generates K63-linked polyubiquitin chains. This modified substrate is subsequently recognized by RNF168 and tethers the protein to the DNA lesion. RNF168 then ubiquitinates proteins such as histone H2A and H2AX to further amplify the damage response and recruit repair proteins such as BRCA1 and 53BP1 (Figure 1).
- Published
- 2018
33. USP13 regulates the RAP80-BRCA1 complex dependent DNA damage response
- Author
-
Jian Yuan, Zhenkun Lou, Yujiao Yin, Lei Li, Somaira Nowsheen, Chenming Wu, Kuntian Luo, Min Deng, Yunhui Li, and Yuping Chen
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,DNA Repair ,DNA damage ,DNA repair ,Science ,General Physics and Astronomy ,Mice, Nude ,Ataxia Telangiectasia Mutated Proteins ,General Biochemistry, Genetics and Molecular Biology ,Piperazines ,Article ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Endopeptidases ,Transcriptional regulation ,medicine ,Animals ,Humans ,Histone Chaperones ,Phosphorylation ,Cisplatin ,Ovarian Neoplasms ,Multidisciplinary ,Chemistry ,BRCA1 Protein ,Ubiquitination ,Nuclear Proteins ,General Chemistry ,Xenograft Model Antitumor Assays ,Protein ubiquitination ,DNA-Binding Proteins ,030104 developmental biology ,Drug Resistance, Neoplasm ,Multiprotein Complexes ,PARP inhibitor ,Cancer research ,Phthalazines ,Female ,Ubiquitin-Specific Proteases ,Carrier Proteins ,medicine.drug - Abstract
BRCA1 regulates multiple cellular pathways that maintain genomic stability including cell cycle checkpoints, DNA repair, protein ubiquitination, chromatin remodelling, transcriptional regulation and apoptosis. Receptor-associated protein 80 (RAP80) helps recruit BRCA1 to double-strand breaks (DSBs) through the scaffold protein CCDC98 (Abraxas) and facilitates DNA damage response (DDR). However, the regulation of RAP80-BRCA1 complex is still unclear. Here we report that a deubiquitinase, USP13, regulates DDR by targeting RAP80. Mechanistically, USP13 is phosphorylated by ATM following DNA damage which, in turn, facilitates its DSB localization. USP13, in turn, deubiquitinates RAP80 and promotes RAP80 recruitment and proper DDR. Depleting or inhibiting USP13 sensitizes ovarian cancer cells to cisplatin and PARP inhibitor (olaparib) while overexpression of USP13 renders ovarian cancer cells resistant to chemotherapy. Overall, we identify USP13 as a regulator of DNA repair and reveal a model in which a phosphorylation-deubiquitination axis dynamically regulates RAP80-BRCA1 complex foci formation and function., RAP80 helps to recruit BRCA1 to double-strand breaks, facilitating DNA damage responses. Here the authors report that phosphorylated USP13 deubiquitinates RAP80 after DNA damage, prompting recruitment to the break site.
- Published
- 2017
34. USP49 negatively regulates tumorigenesis and chemoresistance through FKBP51‐AKT signaling
- Author
-
Jian Yuan, Chenming Wu, Kuntian Luo, Somaira Nowsheen, Zhenkun Lou, Lizhi Zhang, Qi Hu, Lei Li, Yunhui Li, Yujiao Yin, and Yuping Chen
- Subjects
0301 basic medicine ,Scaffold protein ,Antimetabolites, Antineoplastic ,Carcinogenesis ,Cell Survival ,Drug Resistance ,Regulator ,Biology ,medicine.disease_cause ,Deoxycytidine ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Tacrolimus Binding Proteins ,03 medical and health sciences ,0302 clinical medicine ,Pancreatic cancer ,Phosphoprotein Phosphatases ,medicine ,Humans ,Phosphorylation ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,General Immunology and Microbiology ,Cell growth ,General Neuroscience ,Nuclear Proteins ,Articles ,medicine.disease ,Gemcitabine ,Cell biology ,Pancreatic Neoplasms ,030104 developmental biology ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,Signal transduction ,Protein Processing, Post-Translational ,Proto-Oncogene Proteins c-akt ,Ubiquitin Thiolesterase ,Signal Transduction - Abstract
The AKT pathway is a fundamental signaling pathway that mediates multiple cellular processes, such as cell proliferation and survival, angiogenesis, and glucose metabolism. We recently reported that the immunophilin FKBP51 is a scaffolding protein that can enhance PHLPP‐AKT interaction and facilitate PHLPP‐mediated dephosphorylation of AKT at Ser473, negatively regulating AKT activation. However, the regulation of FKBP51‐PHLPP‐AKT pathway remains unclear. Here we report that a deubiquitinase, USP49, is a new regulator of the AKT pathway. Mechanistically, USP49 deubiquitinates and stabilizes FKBP51, which in turn enhances PHLPP9s capability to dephosphorylate AKT. Furthermore, USP49 inhibited pancreatic cancer cell proliferation and enhanced cellular response to gemcitabine in a FKBP51‐AKT‐dependent manner. Clinically, decreased expression of USP49 in patients with pancreatic cancer was associated with decreased FKBP51 expression and increased AKT phosphorylation. Overall, our findings establish USP49 as a novel regulator of AKT pathway with a critical role in tumorigenesis and chemo‐response in pancreatic cancer.
- Published
- 2017
35. CDK4/6-dependent activation of DUB3 regulates cancer metastasis through SNAIL1
- Author
-
Jia Yu, Tao Ren, Haoxing Zhang, Matthew P. Goetz, Chenming Wu, Jung Jin Kim, Yang Han, Yunhui Li, Peng Yin, Seung Baek Lee, Kuntian Luo, Zhenkun Lou, Lizhi Zhang, Bo Qin, Yujiao Yin, Tongzheng Liu, Liewei Wang, Judy C. Boughey, Min Deng, Somaira Nowsheen, Jian Yuan, Jun Zhang, and Jing Lin
- Subjects
0301 basic medicine ,Lung Neoplasms ,Leupeptins ,Pyridines ,General Physics and Astronomy ,Triple Negative Breast Neoplasms ,Piperazines ,Deubiquitinating enzyme ,Mice ,Cell Movement ,Medicine ,RNA, Small Interfering ,Ovarian Neoplasms ,Regulation of gene expression ,Tumour metastasis ,Multidisciplinary ,biology ,Liver Neoplasms ,Tumour site ,3. Good health ,Gene Expression Regulation, Neoplastic ,MCF-7 Cells ,Female ,Signal transduction ,Signal Transduction ,Science ,Cancer metastasis ,Antineoplastic Agents ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cell Line, Tumor ,Endopeptidases ,Animals ,Humans ,business.industry ,Cyclin-Dependent Kinase 4 ,Breast cancer metastasis ,Cyclin-Dependent Kinase 6 ,General Chemistry ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,Cancer cell ,Immunology ,Cancer research ,biology.protein ,Snail Family Transcription Factors ,business - Abstract
Tumour metastasis, the spread of cancer cells from the original tumour site followed by growth of secondary tumours at distant organs, is the primary cause of cancer-related deaths and remains poorly understood. Here we demonstrate that inhibition of CDK4/6 blocks breast tumour metastasis in the triple-negative breast cancer model, without affecting tumour growth. Mechanistically, we identify a deubiquitinase, DUB3, as a target of CDK4/6; CDK4/6-mediated activation of DUB3 is essential to deubiquitinate and stabilize SNAIL1, a key factor promoting epithelial–mesenchymal transition and breast cancer metastasis. Overall, our study establishes the CDK4/6–DUB3 axis as an important regulatory mechanism of breast cancer metastasis and provides a rationale for potential therapeutic interventions in the treatment of breast cancer metastasis., Overexpression of SNAIL confers tumour cells with cancer stem-like characteristics associated with tumour progression. Here the authors show that inhibition of CDK4/6 blocks tumour metastasis in triple negative breast cancer by targeting DUB3 which in turns deubiquitinates and stabilises SNAIL1.
- Published
- 2017
36. A phosphorylation–deubiquitination cascade regulates the BRCA2–RAD51 axis in homologous recombination
- Author
-
Lei Li, Zhenkun Lou, Kuntian Luo, Somaira Nowsheen, Yuping Chen, Yujiao Yin, Jian Yuan, Chenming Wu, Yunhui Li, and Min Deng
- Subjects
0301 basic medicine ,Genome instability ,DNA Repair ,DNA damage ,DNA repair ,genetic processes ,RAD51 ,Breast Neoplasms ,Poly(ADP-ribose) Polymerase Inhibitors ,Radiation Tolerance ,Gene Knockout Techniques ,03 medical and health sciences ,Ubiquitin ,Cell Line, Tumor ,Genetics ,Humans ,Phosphorylation ,Homologous Recombination ,skin and connective tissue diseases ,BRCA2 Protein ,biology ,Ubiquitination ,Survival Analysis ,Cell biology ,Gene Expression Regulation, Neoplastic ,Non-homologous end joining ,Cysteine Endopeptidases ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,030104 developmental biology ,MCF-7 Cells ,health occupations ,biology.protein ,Rad51 Recombinase ,biological phenomena, cell phenomena, and immunity ,Homologous recombination ,Ubiquitin Thiolesterase ,Protein Binding ,Signal Transduction ,Research Paper ,Developmental Biology ,Deubiquitination - Abstract
Homologous recombination (HR) is one of the major DNA double-strand break (DSB) repair pathways in mammalian cells. Defects in HR trigger genomic instability and result in cancer predisposition. The defining step of HR is homologous strand exchange directed by the protein RAD51, which is recruited to DSBs by BRCA2. However, the regulation of the BRCA2–RAD51 axis remains unclear. Here we report that ubiquitination of RAD51 hinders RAD51–BRCA2 interaction, while deubiquitination of RAD51 facilitates RAD51–BRCA2 binding and RAD51 recruitment and thus is critical for proper HR. Mechanistically, in response to DNA damage, the deubiquitinase UCHL3 is phosphorylated and activated by ATM. UCHL3, in turn, deubiquitinates RAD51 and promotes the binding between RAD51 and BRCA2. Overexpression of UCHL3 renders breast cancer cells resistant to radiation and chemotherapy, while depletion of UCHL3 sensitizes cells to these treatments, suggesting a determinant role of UCHL3 in cancer therapy. Overall, we identify UCHL3 as a novel regulator of DNA repair and reveal a model in which a phosphorylation–deubiquitination cascade dynamically regulates the BRCA2–RAD51 pathway.
- Published
- 2016
37. The bromodomain containing protein BRD-9 orchestrates RAD51-RAD54 complex formation and regulates homologous recombination-mediated repair
- Author
-
Chao Zhang, Qin Zhou, Jinzhou Huang, Yong Zhang, Baohua Liu, Somaira Nowsheen, Xinyi Tu, Bo Qin, Yuping Chen, Qian Zhu, Wootae Kim, Min Deng, Jian Yuan, Fei Zhao, Zhenkun Lou, Kuntian Luo, and Robert W. Mutter
- Subjects
0301 basic medicine ,Molecular biology ,RAD51 ,General Physics and Astronomy ,Synthetic lethality ,Biochemistry ,Piperazines ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,lcsh:Science ,Ovarian Neoplasms ,Multidisciplinary ,DNA-Binding Proteins ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Heterografts ,Female ,Cell biology ,DNA damage ,Science ,Mice, Nude ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,Article ,Genomic Instability ,Olaparib ,03 medical and health sciences ,Cell Line, Tumor ,Animals ,Humans ,Protein Interaction Domains and Motifs ,fungi ,DNA Helicases ,Recombinational DNA Repair ,General Chemistry ,Bromodomain ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,Multiprotein Complexes ,Cancer cell ,Cancer research ,Phthalazines ,lcsh:Q ,Rad51 Recombinase ,Cisplatin ,Homologous recombination ,Transcription Factors - Abstract
Homologous recombination (HR) is important for error-free DNA double strand break repair and maintenance of genomic stability. However, upregulated HR is also used by cancer cells to promote therapeutic resistance. Therefore, inducing HR deficiency (HRD) is a viable strategy to sensitize HR proficient cancers to DNA targeted therapies in order to overcome therapeutic resistance. A bromodomain containing protein, BRD9, was previously reported to regulate chromatin remodeling and transcription. Here, we discover that following DNA damage, the bromodomain of BRD9 binds acetylated K515 on RAD54 and facilitates RAD54’s interaction with RAD51, which is essential for HR. BRD9 is overexpressed in ovarian cancer and depleting BRD9 sensitizes cancer cells to olaparib and cisplatin. In addition, inhibitor of BRD9, I-BRD9, acts synergistically with olaparib in HR-proficient cancer cells. Overall, our results elucidate a role for BRD9 in HR and identify BRD9 as a potential therapeutic target to promote synthetic lethality and overcome chemoresistance., The bromodomain containing protein BRD9 has been reported to regulate chromatin remodeling and transcription. Here the authors reveal a role for BRD9 in homologous recombination by facilitating RAD51–RAD54 interaction.
- Published
- 2019
38. ZFP161 regulates replication fork stability and maintenance of genomic stability by recruiting the ATR/ATRIP complex
- Author
-
Zhenkun Lou, Sisi Qin, Kuntian Luo, Fei Zhao, Jian Yuan, Wootae Kim, Min Deng, Qin Zhou, Jinzhou Huang, Somaira Nowsheen, Guijie Guo, Rentian Wu, and Yuping Chen
- Subjects
0301 basic medicine ,Scaffold protein ,Genome instability ,DNA Replication ,DNA damage ,Science ,Kruppel-Like Transcription Factors ,General Physics and Astronomy ,Ataxia Telangiectasia Mutated Proteins ,ATR-ATRIP complex ,General Biochemistry, Genetics and Molecular Biology ,Genomic Instability ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,stomatognathic system ,Replication Protein A ,Animals ,Humans ,lcsh:Science ,Replication protein A ,Adaptor Proteins, Signal Transducing ,Cancer ,Zinc finger ,Mice, Knockout ,Multidisciplinary ,Chemistry ,DNA replication ,Signal transducing adaptor protein ,General Chemistry ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,HEK293 Cells ,030220 oncology & carcinogenesis ,Checkpoint Kinase 1 ,lcsh:Q ,biological phenomena, cell phenomena, and immunity ,DNA Damage ,Signal Transduction - Abstract
DNA replication stress-mediated activation of the ATR kinase pathway is important for maintaining genomic stability. In this study, we identified a zinc finger protein, ZFP161 that functions as a replication stress response factor in ATR activation. Mechanistically, ZFP161 acts as a scaffolding protein to facilitate the interaction between RPA and ATR/ATRIP. ZFP161 binds to RPA and ATR/ATRIP through distinct regions and stabilizes the RPA–ATR–ATRIP complex at stalled replication forks. This function of ZFP161 is important to the ATR signaling cascade and genome stability maintenance. In addition, ZFP161 knockout mice showed a defect in ATR activation and genomic instability. Furthermore, low expression of ZFP161 is associated with higher cancer risk and chromosomal instability. Overall, these findings suggest that ZFP161 coordinates ATR/Chk1 pathway activation and helps maintain genomic stability., The ATR pathway is active during DNA replication stress to maintain genome stability. Here the authors reveal the role of the zinc finger containing protein 161 (ZFP161) to facilitate replication fork stability by acting as a scaffold to facilitate the interaction between RPA and ATR/ATRIP.
- Published
- 2019
39. A novel UCHL3 inhibitor, perifosine, enhances PARP inhibitor cytotoxicity through inhibition of homologous recombination-mediated DNA double strand break repair
- Author
-
Jinzhou Huang, Xinyi Tu, Yuping Chen, Qin Zhou, Zhiwang Song, Jiaqi Liu, Somaira Nowsheen, Seung Baek Lee, Jian Yuan, Zhenkun Lou, Wootae Kim, and Kuntian Luo
- Subjects
0301 basic medicine ,Cancer Research ,lcsh:Cytology ,DNA damage ,Chemistry ,DNA repair ,Immunology ,RAD51 ,Cell Biology ,Perifosine ,3. Good health ,Olaparib ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cancer cell ,PARP inhibitor ,Cancer research ,lcsh:QH573-671 ,Protein kinase B - Abstract
Triple-negative breast cancer (TNBC) treatment remains a great challenge for clinical practice and novel therapeutic strategies are urgently needed. UCHL3 is a deubiquitinase that is overexpressed in TNBC and correlates with poor prognosis. UCHL3 deubiquitinates RAD51 thereby promoting the recruitment of RAD51 to DNA damage sites and augmenting DNA repair. Therefore, UCHL3 overexpression can render cancer cells resistant to DNA damage inducing chemo and radiotherapy, and targeting UCHL3 can sensitize TNBC to radiation and chemotherapy. However, small molecule inhibitors of UCHL3 are yet to be identified. Here we report that perifosine, a previously reported Akt inhibitor, can inhibit UCHL3 in vitro and in vivo. We found low dose (50 nM) perifosine inhibited UCHL3 deubiquitination activity without affecting Akt activity. Furthermore, perifosine enhanced Olaparib-induced growth inhibition in TNBC cells. Mechanistically, perifosine induced RAD51 ubiquitination and blocked the RAD51-BRCA2 interaction, which in turn decreased ionizing radiation-induced foci (IRIF) of Rad51 and, thereby, homologous recombination (HR)-mediated DNA double strand break repair. In addition, combination of perifosine and Olaparib showed synergistic antitumor activity in vivo in TNBC xenograft model. Thus, our present study provides a novel therapeutic approach to optimize PARP inhibitor treatment efficiency.
- Published
- 2019
40. USP39 regulates DNA damage response and chemo-radiation resistance by deubiquitinating and stabilizing CHK2
- Author
-
Jinhuan Wu, Somaira Nowsheen, Wootae Kim, Zhenkun Lou, Jian Yuan, Xinyi Tu, Jiaqi Liu, Haixuan Qiao, Yuping Chen, Guijie Guo, Yunhui Li, Qin Zhou, Xiumei Gao, Chenming Wu, Fei Zhao, Guohe Geng, Ping Yin, Jinzhou Huang, Lei Li, Kuntian Luo, and Chao Zhang
- Subjects
0301 basic medicine ,Cancer Research ,animal structures ,Cell cycle checkpoint ,Lung Neoplasms ,DNA Repair ,DNA repair ,DNA damage ,Regulator ,environment and public health ,Radiation Tolerance ,Small hairpin RNA ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Cell Line, Tumor ,Humans ,Checkpoint Kinase 2 ,biology ,Kinase ,Chemistry ,Protein Stability ,Cell Cycle ,Ubiquitination ,Cell biology ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Oncology ,A549 Cells ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,biology.protein ,Ubiquitin-Specific Proteases ,biological phenomena, cell phenomena, and immunity ,DNA Damage - Abstract
The serine/threonine kinase, CHK2 (checkpoint kinase 2), is a key mediator in DNA damage response and a tumor suppressor, which is implicated in promoting cell cycle arrest, apoptosis and DNA repair. Accumulating evidence suggests that these functions are primarily exerted through phosphorylation downstream factors such as p53 and BRCA1. Recent studies have shown that ubiquitination is an important mode of regulation of CHK2. However, it remains largely unclear whether deubiquitinases participate in regulation of CHK2. Here, we report that a deubiquitinase, USP39, is a new regulator of CHK2. Mechanistically, USP39 deubiquitinates and stabilizes CHK2, which in turn enhances CHK2 stability. Short hairpin RNA (shRNA) mediated knockdown of USP39 led to deregulate CHK2, which resulted in compromising the DNA damage-induced G2/M checkpoint, decreasing apoptosis, and conferring cancer cells resistance to chemotherapy drugs and radiation treatment. Collectively, we identify USP39 as a novel regulator of CHK2 in the DNA damage response.
- Published
- 2018
41. Tandem Deubiquitination and Acetylation of SPRTN Promotes DNA-Protein Crosslink Repair and Protects against Aging
- Author
-
Xinyi Tu, Ming Gao, Guijie Guo, Qian Zhu, Yusuke Kojima, Wootae Kim, Bo Qin, Qin Zhou, Ping Yin, Min Deng, Yong Zhang, Jinzhou Huang, Somaira Nowsheen, Yuichi J. Machida, Fei Zhao, Zhenkun Lou, and Kuntian Luo
- Subjects
Male ,Genome instability ,Premature aging ,Aging ,DNA Repair ,DNA repair ,Genomic Instability ,Article ,Cell Line ,Deubiquitinating enzyme ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Ubiquitin ,Endopeptidases ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Deubiquitinating Enzymes ,biology ,Ubiquitination ,Acetylation ,Cell Biology ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,HEK293 Cells ,biology.protein ,Female ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery ,DNA Damage ,Deubiquitination - Abstract
DNA-protein crosslinks (DPCs) are highly toxic DNA lesions that threaten genomic integrity. Recent findings highlight that SPRTN, a specialized DNA-dependent metalloprotease, is a central player in proteolytic cleavage of DPCs. Previous studies suggest that SPRTN deubiquitination is important for its chromatin association and activation. However, the regulation and consequence of SPRTN deubiquitination remain unclear. Here, we report that in response to DPC induction, the deubiquitinase VCPIP1/VCIP135 is phosphorylated and activated by ATM/ATR. VCPIP1, in turn, deubiquitinates SPRTN and promotes its chromatin relocalization. The deubiquitination of SPRTN is required for its subsequent acetylation, which promotes SPRTN relocation to the site of chromatin damage. Furthermore, Vcpip1 knockout mice are prone to genomic instability and premature aging. We propose a model that two sequential post-translational modifications (PTMs) regulate SPRTN chromatin accessibility to repair DPCs and maintain genomic stability and healthy life span.
- Published
- 2020
42. A novel topoisomerase I inhibitor DIA-001 induces DNA damage mediated cell cycle arrest and apoptosis in cancer cell
- Author
-
Shiguang Zhao, Kuntian Luo, Guohe Geng, Jiaqi Liu, Ling Wang, Jian Yuan, and Guang Liang
- Subjects
Cell cycle checkpoint ,biology ,DNA damage ,Topoisomerase ,DNA replication ,General Medicine ,Cell cycle ,Topoisomerase-I Inhibitor ,Molecular biology ,chemistry.chemical_compound ,chemistry ,Cancer cell ,biology.protein ,Original Article ,DNA - Abstract
Background DNA topoisomerase enzyme plays an essential role in controlling the DNA topology structure by binding to DNA and cutting the phosphate backbone of either one or both of the DNA strands. Here, we have identified a small molecule inhibitor, DIA-001, that directly binds to Topoisomerase 1 (Topo I) and promotes the Topo I-DNA adducts. Methods In this study, we investigated the antitumor effects of DIA-001 using MTS assay and colony formation. We examined cell cycle of tumor cells with DIA-001 treatment in vitro by flow cytometry. And we investigated DNA damage and cell cycle marker protein after treatment with DIA-001 at different concentration and time point by western blot. Immunofluorescence was performance to detect the nuclear foci. The effects of DIA-001 on Topo I and Topo II activities were examined by DNA relaxation assays. Results We demonstrate that DIA-001 inhibit DNA replication and arrest cell cycle progression at the G2/M phase by directly binds to Topo I and promotes the Topo I-DNA adducts. In addition, DIA-001 can activate the DNA damage response signaling cascade, resulting in apoptosis in treated cells. Conclusions Our findings show a novel compound for treatment of cancer cells with the potential as a chemotherapy candidate that is less toxic to normal cells.
- Published
- 2020
43. A novel UCHL
- Author
-
Zhiwang, Song, Xinyi, Tu, Qin, Zhou, Jinzhou, Huang, Yuping, Chen, Jiaqi, Liu, SeungBaek, Lee, Wootae, Kim, Somaira, Nowsheen, Kuntian, Luo, Jian, Yuan, and Zhenkun, Lou
- Subjects
BRCA2 Protein ,Phosphorylcholine ,Transplantation, Heterologous ,Ubiquitination ,Mice, Nude ,Recombinational DNA Repair ,Antineoplastic Agents ,Apoptosis ,Triple Negative Breast Neoplasms ,Poly(ADP-ribose) Polymerase Inhibitors ,Piperazines ,Article ,Mice ,Cell growth ,Cancer therapeutic resistance ,Cell Line, Tumor ,Radiation, Ionizing ,Animals ,Humans ,Phthalazines ,Female ,Rad51 Recombinase ,Ubiquitin Thiolesterase ,RNA, Guide, Kinetoplastida - Abstract
Triple-negative breast cancer (TNBC) treatment remains a great challenge for clinical practice and novel therapeutic strategies are urgently needed. UCHL3 is a deubiquitinase that is overexpressed in TNBC and correlates with poor prognosis. UCHL3 deubiquitinates RAD51 thereby promoting the recruitment of RAD51 to DNA damage sites and augmenting DNA repair. Therefore, UCHL3 overexpression can render cancer cells resistant to DNA damage inducing chemo and radiotherapy, and targeting UCHL3 can sensitize TNBC to radiation and chemotherapy. However, small molecule inhibitors of UCHL3 are yet to be identified. Here we report that perifosine, a previously reported Akt inhibitor, can inhibit UCHL3 in vitro and in vivo. We found low dose (50 nM) perifosine inhibited UCHL3 deubiquitination activity without affecting Akt activity. Furthermore, perifosine enhanced Olaparib-induced growth inhibition in TNBC cells. Mechanistically, perifosine induced RAD51 ubiquitination and blocked the RAD51-BRCA2 interaction, which in turn decreased ionizing radiation-induced foci (IRIF) of Rad51 and, thereby, homologous recombination (HR)-mediated DNA double strand break repair. In addition, combination of perifosine and Olaparib showed synergistic antitumor activity in vivo in TNBC xenograft model. Thus, our present study provides a novel therapeutic approach to optimize PARP inhibitor treatment efficiency.
- Published
- 2018
44. Chk1 inhibitor SCH 900776 enhances the antitumor activity of MLN4924 on pancreatic cancer
- Author
-
Jian Yuan, Zhenkun Lou, Jian ang Li, Chao Song, Yefei Rong, Tiantao Kuang, Somaira Nowsheen, Dansong Wang, Xuefeng Xu, Bo Qin, Kuntian Luo, and Wenhui Lou
- Subjects
0301 basic medicine ,Adult ,DNA Replication ,Male ,Combination therapy ,DNA damage ,medicine.medical_treatment ,Mice, Nude ,Cyclopentanes ,Ubiquitin-Activating Enzymes ,Biology ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Pancreatic cancer ,Report ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Cytotoxicity ,Molecular Biology ,Protein Kinase Inhibitors ,Aged ,Aged, 80 and over ,Drug Synergism ,Cell Biology ,Cell cycle ,Middle Aged ,medicine.disease ,Prognosis ,G2 Phase Cell Cycle Checkpoints ,Pancreatic Neoplasms ,030104 developmental biology ,Pyrimidines ,Proteasome ,030220 oncology & carcinogenesis ,Checkpoint Kinase 1 ,Cancer research ,Pyrazoles ,Female ,Developmental Biology ,DNA Damage - Abstract
MLN4924 inhibits the cullin-RING ligases mediated ubiquitin-proteasome system, and has showed antitumor activities in preclinical studies, but its effects and mechanisms on pancreatic cancer (PC) remains elusive. We found that MLN4924 inhibited the proliferation and clonogenicity of PC cells, caused DNA damage, particularly double-strand breaks, and leaded to Chk1 activation and cell-cycle arrest. Chk1 inhibitor SCH 900776 alone exhibited minimal cytotoxicity, and caused no DNA damage on PC cells. But in the combination therapy, SCH 900776 enhanced the cytotoxicity and DNA damage caused by MLN4924, likely by abrogating G2/M arrest and promoting DNA re-replication. In vivo study on a xenograft PC mouse model also showed that SCH 900776 increased the efficacy of MLN4924. We also evaluated the level of NEDD8-activating enzyme (NAE), the direct target of MLN4924, and found that NAE level was elevated in PC tissues compared with normal pancreas, but was irrelevant with prognosis. Our findings provide the preclinical evidence and the rationale of the combination therapy of MLN4924 with SCH 900776 or other Chk1 inhibitors to treat PC.
- Published
- 2018
45. A Divergent Role of the SIRT1-TopBP1 Axis in Regulating Metabolic Checkpoint and DNA Damage Checkpoint
- Author
-
Sung Yun Jung, Yunhui Li, Leng Wenchuan, Jun Qin, Bo Qin, Zhenkun Lou, Tongzheng Liu, Yi Hui Lin, Kuntian Luo, Jian Yuan, Min Deng, Haoxing Zhang, and Debra Evans
- Subjects
DNA Replication ,Cell cycle checkpoint ,DNA Repair ,DNA repair ,DNA damage ,Protein Conformation ,Cell Cycle Proteins ,Biology ,Article ,Cell Line ,chemistry.chemical_compound ,Mice ,Sirtuin 1 ,Stress, Physiological ,Animals ,Humans ,CHEK1 ,Molecular Biology ,DNA replication ,Acetylation ,Cell Cycle Checkpoints ,Cell Biology ,G2-M DNA damage checkpoint ,Cell biology ,HEK293 Cells ,DNA Replication Inhibition ,chemistry ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins ,DNA ,DNA Damage ,Signal Transduction - Abstract
DNA replication is executed only when cells have sufficient metabolic resources and undamaged DNA. Nutrient limitation and DNA damage cause a metabolic checkpoint and DNA damage checkpoint, respectively. Although SIRT1 activity is regulated by metabolic stress and DNA damage, its function in these stress-mediated checkpoints remains elusive. Here we report that the SIRT1-TopBP1 axis functions as a switch for both checkpoints. With glucose deprivation, SIRT1 is activated and deacetylates TopBP1, resulting in TopBP1-Treslin disassociation and DNA replication inhibition. Conversely, SIRT1 activity is inhibited under genotoxic stress, resulting in increased TopBP1 acetylation that is important for the TopBP1-Rad9 interaction and activation of the ATR-Chk1 pathway. Mechanistically, we showed that acetylation of TopBP1 changes the conformation of TopBP1, thereby facilitating its interaction with distinct partners in DNA replication and checkpoint activation. Taken together, our studies identify the SIRT1-TopBP1 axis as a key signaling mode in the regulation of the metabolic checkpoint and the DNA damage checkpoint.
- Published
- 2014
- Full Text
- View/download PDF
46. The deubiquitinase USP9X promotes tumor cell survival and confers chemoresistance through YAP1 stabilization
- Author
-
Kuntian Luo, Zhenkun Lou, Chenming Wu, Tongzheng Liu, Yunhui Li, Yujiao Yin, Jinhuan Wu, Jian Yuan, Yuping Chen, Lei Li, and Somaira Nowsheen
- Subjects
0301 basic medicine ,Cancer Research ,Cell Survival ,Mice, Nude ,Breast Neoplasms ,Biology ,Article ,03 medical and health sciences ,Mice ,Breast cancer ,Antineoplastic Combined Chemotherapy Protocols ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Transcription factor ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,YAP1 ,Hippo signaling pathway ,Effector ,Protein Stability ,Cancer ,YAP-Signaling Proteins ,medicine.disease ,Phosphoproteins ,3. Good health ,030104 developmental biology ,USP9X ,HEK293 Cells ,Drug Resistance, Neoplasm ,Cancer cell ,Cancer research ,MCF-7 Cells ,Female ,Protein Processing, Post-Translational ,Ubiquitin Thiolesterase ,Transcription Factors - Abstract
The Yes-associated protein 1 (YAP1), a major downstream effector of the Hippo pathway, functions as a transcriptional regulator and has an important role in cellular control of organ size and tumor growth. Elevated oncogenic activity of YAP1 has been clarified in different types of human cancers, which contributes to cancer cell survival and chemoresistance. However, the molecular mechanism of YAP1 overexpression in cancer is still not clear. Here we demonstrate that the deubiquitination enzyme USP9X deubiquitinates and stabilizes YAP1, thereby promoting cancer cell survival. Increased USP9X expression correlates with increased YAP1 protein in human breast cancer cell lines and patient samples. Moreover, depletion of USP9X increases YAP1 polyubiquitination, which in turn elevates YAP1 turnover and cell sensitivity to chemotherapy. Overall, our study establishes the USP9X-YAP1 axis as an important regulatory mechanism of breast cancer and provides a rationale for potential therapeutic interventions in the treatment of breast cancer.
- Published
- 2017
47. HERC2-USP20 axis regulates DNA damage checkpoint through Claspin
- Author
-
Zhenkun Lou, Ping Yin, Bowen Gao, Min Deng, Puqiang Wu, Jian Yuan, Tongzheng Liu, Yuan Fang, Yunhui Li, and Kuntian Luo
- Subjects
DNA Replication ,Cell cycle checkpoint ,DNA Repair ,DNA damage ,DNA repair ,Ubiquitin-Protein Ligases ,Mice, Nude ,Ataxia Telangiectasia Mutated Proteins ,Genome Integrity, Repair and Replication ,Biology ,Cell Line ,Mice ,Endopeptidases ,Genetics ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Phosphorylation ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Tumor Suppressor Proteins ,DNA replication ,Cell Cycle Checkpoints ,G2-M DNA damage checkpoint ,3. Good health ,Ubiquitin ligase ,HEK293 Cells ,biology.protein ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Ubiquitin Thiolesterase ,DNA Damage ,Deubiquitination - Abstract
The DNA damage response triggers cell-cycle checkpoints, DNA repair and apoptosis using multiple post-translational modifications as molecular switches. However, how ubiquitination regulates ATR signaling in response to replication stress and single-strand break is still unclear. Here, we identified the deubiquitination enzyme (DUB) USP20 as a pivotal regulator of ATR-related DDR pathway. Through screening a panel of DUBs, we identified USP20 as critical for replication stress response. USP20 is phosphorylated by ATR, resulting in disassociation of the E3 ubiquitin ligase HERC2 from USP20 and USP20 stabilization. USP20 in turn deubiquitinates and stabilizes Claspin and enhances the activation of ATR-Chk1 signaling. These findings reveal USP20 to be a novel regulator of ATR-dependent DNA damage signaling.
- Published
- 2014
48. Author Correction: Topoisomerase IIα controls the decatenation checkpoint
- Author
-
Junjie Chen, Jian Yuan, Kuntian Luo, and Zhenkun Lou
- Subjects
Blot ,Topoisomerase iiα ,Chemistry ,Nat ,Cell Biology ,Molecular biology ,Cell biology - Abstract
In Fig. 1b of this Article, the authors mistakenly republished a blot previously published as Fig. 1d in their article in Nature Structural & Molecular Biology (Nat. Struct. Mol. Biol. 12, 589-593; 2005).
- Published
- 2018
49. Sumoylation of MDC1 is important for proper DNA damage response
- Author
-
Haoxing Zhang, Kuntian Luo, Liewei Wang, Jian Yuan, and Zhenkun Lou
- Subjects
Mutation ,General Immunology and Microbiology ,biology ,DNA damage ,DNA repair ,General Neuroscience ,medicine.disease_cause ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology ,Proliferating cell nuclear antigen ,MDC1 ,biology.protein ,medicine ,DNA mismatch repair ,Homologous recombination ,Molecular Biology ,Replication protein A - Abstract
In response to DNA damage, many DNA damage factors, such as MDC1 and 53BP1, redistribute to sites of DNA damage. The mechanism governing the turnover of these factors at DNA damage sites, however, remains enigmatic. Here, we show that MDC1 is sumoylated following DNA damage, and the sumoylation of MDC1 at Lys1840 is required for MDC1 degradation and removal of MDC1 and 53BP1 from sites of DNA damage. Sumoylated MDC1 is recognized and ubiquitinated by the SUMO-targeted E3 ubiquitin ligase RNF4. Mutation of the MDC1 Lys 1840 (K1840R) results in impaired CtIP, replication protein A, and Rad51 accumulation at sites of DNA damage and defective homologous recombination (HR). The HR defect caused by MDC1K1840R mutation could be rescued by 53BP1 downregulation. These results reveal the intricate dynamics governing the assembly and disassembly of DNA damage factors at sites of DNA damage for prompt response to DNA damage.
- Published
- 2012
50. MMSET regulates histone H4K20 methylation and 53BP1 accumulation at DNA damage sites
- Author
-
Zhenkun Lou, Huadong Pei, Marta Chesi, Zhongsheng You, Lindsey M Zhang, Kuntian Luo, P. Leif Bergsagel, Frances Fei, Yuxin Qin, and Liewei Wang
- Subjects
Chromatin Immunoprecipitation ,DNA damage ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Methylation ,Article ,Histone H4 ,Histones ,03 medical and health sciences ,Phosphoserine ,0302 clinical medicine ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,0303 health sciences ,Multidisciplinary ,biology ,Lysine ,Tumor Suppressor Proteins ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Histone-Lysine N-Methyltransferase ,Molecular biology ,MDC1 ,DNA-Binding Proteins ,Repressor Proteins ,Protein Transport ,BRCT domain ,Histone ,HEK293 Cells ,030220 oncology & carcinogenesis ,Histone methyltransferase ,biology.protein ,Trans-Activators ,Tumor Suppressor p53-Binding Protein 1 ,Chromatin immunoprecipitation ,HeLa Cells - Abstract
p53-binding protein 1 (53BP1) is known to be an important mediator of the DNA damage response, with dimethylation of histone H4 lysine 20 (H4K20me2) critical to the recruitment of 53BP1 to double-strand breaks (DSBs). However, it is not clear how 53BP1 is specifically targeted to the sites of DNA damage, as the overall level of H4K20me2 does not seem to increase following DNA damage. It has been proposed that DNA breaks may cause exposure of methylated H4K20 previously buried within the chromosome; however, experimental evidence for such a model is lacking. Here we found that H4K20 methylation actually increases locally upon the induction of DSBs and that methylation of H4K20 at DSBs is mediated by the histone methyltransferase MMSET (also known as NSD2 or WHSC1) in mammals. Downregulation of MMSET significantly decreases H4K20 methylation at DSBs and the subsequent accumulation of 53BP1. Furthermore, we found that the recruitment of MMSET to DSBs requires the γH2AX-MDC1 pathway; specifically, the interaction between the MDC1 BRCT domain and phosphorylated Ser 102 of MMSET. Thus, we propose that a pathway involving γH2AX-MDC1-MMSET regulates the induction of H4K20 methylation on histones around DSBs, which, in turn, facilitates 53BP1 recruitment.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.